1
|
Elhaieg A, Farag A, Koung Ngeun S, Kaneda M, Yokoi A, Mandour AS, Tanaka R. Therapeutic Potential of Local and Systemic Adipose-Derived Mesenchymal Stem Cell Injections in a Rat Model of Experimental Periodontitis: Implications for Cardiac Function. Int J Mol Sci 2025; 26:3984. [PMID: 40362223 PMCID: PMC12071214 DOI: 10.3390/ijms26093984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/10/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Periodontitis is a common inflammatory disease that not only damages periodontal tissues but also induces systemic effects, including cardiac dysfunction. Mesenchymal stem cells (MSCs) offer regenerative potential due to their ability to differentiate, modulate immune responses, and secrete anti-inflammatory factors. However, the relative efficacy of local versus systemic MSC administration remains unclear. This study evaluated the therapeutic effects of adipose-derived MSCs (AD-MSCs) in a rat model of experimental periodontitis, comparing local and systemic administration. AD-MSCs were characterized based on morphology, surface marker expression, and differentiation potential. Ligature-induced periodontitis was established over 60 days, after which AD-MSCs (1 × 106 cells) were administered either supraperiosteally (local group) or intravenously (systemic group). Periodontal regeneration was assessed through clinical, radiographic, and histopathological analyses, while cardiac function was evaluated using echocardiography and histopathological examinations. Results demonstrated that local AD-MSC administration provided superior therapeutic benefits compared to systemic delivery. Locally administered cells significantly enhanced bone regeneration, reduced inflammation, and improved periodontal tissue architecture. In contrast, systemic administration offered moderate benefits but was less effective in restoring periodontal integrity. Similarly, in the heart, local treatment resulted in greater improvements in systolic function, as indicated by enhanced ejection fraction and fractional shortening, along with reduced myocardial fibrosis. Although systemic administration also provided cardioprotective effects, diastolic dysfunction persisted in both treatment groups. In conclusion, local AD-MSC administration proved more effective in regenerating periodontal tissues and mitigating cardiac dysfunction, highlighting its potential as an optimized therapeutic strategy for periodontitis and its systemic complications.
Collapse
Affiliation(s)
- Asmaa Elhaieg
- Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (A.F.)
| | - Ahmed Farag
- Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (A.F.)
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Sai Koung Ngeun
- Department of Neurophysiology, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Aimi Yokoi
- Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (A.F.)
| | - Ahmed S. Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ryou Tanaka
- Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan; (A.F.)
| |
Collapse
|
2
|
Zielinska D, Micka-Michalak K, Ademi H, Fisch P, Boeni R, Linder T, Moehrlen U, Biedermann T, Klar AS. Adipose-mesenchymal stem cells enhance the formation of auricular cartilage in vitro and in vivo. Stem Cells Transl Med 2025; 14:szae098. [PMID: 40304399 PMCID: PMC12042163 DOI: 10.1093/stcltm/szae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/11/2024] [Indexed: 05/02/2025] Open
Abstract
Patients suffering from microtia have limited treatment options for auricular reconstruction due to donor-site morbidity, complications, and unaesthetic outcome. Therefore, tissue engineering emerged as an alternative therapeutic option. Here, we generated and characterized human auricular cartilage using differentiated human adipose mesenchymal stem cells (hASCs) combined with human auricular chondrocytes. The differentiated hASCs were analysed for their morphology, phenotype, gene, and protein expression of chondrogenic markers, and biochemical composition at different time points in 2D and 3D in vitro. Importantly, we improved conditions for chondrogenic differentiation of hASCs in vitro to enhance their proliferation, survival, and deposition of cartilaginous-matrix proteins. In particular, gene expression analysis revealed an upregulation of cartilage oligomeric matrix protein (COMP) and aggrecan core protein (ACAN) in hASCs using the improved differentiation protocol in vitro. Additionally, we observed that co-seeding of hASCs with chondrocytes in a 1:5 ratio significantly enhanced the de novo auricular cartilage formation in a collagen-I bioink after 8 weeks on immunodeficient rat. In particular, the co-culture resulted in reduced shrinkage, and increased cartilage matrix production as confirmed by GAG deposition in vivo. Our results demonstrate that in co-cultures, hASCs stimulate cartilage formation due to a synergistic effect: hASCs' differentiation into chondrocytes and a trophic effect of hASCs on human auricular chondrocytes. Here we demonstrate the successful use of an hASC-chondrocyte co-culture technique for auricular cartilage tissue engineering in 3D collagen-I bioink. This co-culture approach omits the major drawbacks of traditional cartilage transplantation and thus, represents a fundamental step towards clinical translation.
Collapse
Affiliation(s)
- Dominika Zielinska
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Katarzyna Micka-Michalak
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Hyrije Ademi
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Philip Fisch
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Roland Boeni
- White House Centre for Liposuction, 8044 Zurich, Switzerland
| | - Thomas Linder
- Otorhinolaryngology Clinic, Cantonal Hospital Luzern, 6000 Luzern, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
- Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, 8008 Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| |
Collapse
|
3
|
Tan MI, Alfarafisa NM, Feronytha AG, Ardisasmita AI, Barlian A, Septiani P. Enhancing exosome-mediated angiotensin-converting enzyme 2 expression modulation: activation of human Wharton's jelly mesenchymal stem cells derived exosomes through curcumin and quinine. Mol Biol Rep 2025; 52:352. [PMID: 40167868 DOI: 10.1007/s11033-025-10454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Cell-free therapy utilizes components such as secretomes and exosomes whose properties depend on the cellular environment. This study investigates how altering the microenvironment of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) with curcumin and quinine affects exosome content, potentially influencing ACE2 expression in Vero and Caco-2 cells. METHODS AND RESULTS hWJ-MSCs were divided into groups treated with curcumin, quinine, DMSO or culture medium only. Secretomes and exosomes were isolated and analyzed, with exosomes characterized by CD63 marker, transmission electron microscopy (TEM), and nanoparticle tracking analysis (NTA). Exosome uptake by Vero and Caco-2 cells was monitored over time, and ACE2 expression was assessed by immunocytochemistry (ICC) and Western blotting. The results of this study demonstrated that exosomes averaged 114.3 nm in size and contained 88 miRNAs, with significant miRNA reductions in exosomes treated with curcumin and quinine. Vero and Caco-2 cells internalized these exosomes within 2.5 h and showed a significant decrease in ACE2 expression when treated with curcumin- or quinine-treated exosomes. This effect was associated with changes in miRNA, including miR-125b-5p in treated exosomes. Exosomes modified with curcumin or quinine significantly downregulated ACE2 expression, suggesting that these compounds may affect miRNA expression or loading, thereby affecting ACE2 protein expression. CONCLUSION This study highlights the therapeutic potential of bioactive compounds in modulating ACE2 through exosome-carried biomolecules. These findings suggest a potential therapeutic strategy for condition associated with ACE2 dysregulation and warrant further investigation into molecular mechanism involved.
Collapse
Affiliation(s)
- Marselina Irasonia Tan
- Department of Biotechnology, School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung, 40132, Indonesia.
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, Sumedang, 45363, Indonesia
| | - Alfina Gracia Feronytha
- Department of Biotechnology, School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung, 40132, Indonesia
| | - Arif Ibrahim Ardisasmita
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Anggraini Barlian
- Department of Biotechnology, School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung, 40132, Indonesia
| | - Popi Septiani
- Department of Biotechnology, School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung, 40132, Indonesia
| |
Collapse
|
4
|
Jammes M, Tabasi A, Bach T, Ritter T. Healing the cornea: Exploring the therapeutic solutions offered by MSCs and MSC-derived EVs. Prog Retin Eye Res 2025; 105:101325. [PMID: 39709150 DOI: 10.1016/j.preteyeres.2024.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Affecting a large proportion of the population worldwide, corneal disorders constitute a concerning health hazard associated to compromised eyesight or blindness for most severe cases. In the last decades, mesenchymal stem/stromal cells (MSCs) demonstrated promising abilities in improving symptoms associated to corneal diseases or alleviating these affections, especially through their anti-inflammatory, immunomodulatory and pro-regenerative properties. More recently, MSC therapeutic potential was shown to be mediated by the molecules they release, and particularly by their extracellular vesicles (EVs; MSC-EVs). Consequently, using MSC-EVs emerged as a pioneering strategy to mitigate the risks related to cell therapy while providing MSC therapeutic benefits. Despite the promises given by MSC- and MSC-EV-based approaches, many improvements are considered to optimize the therapeutic significance of these therapies. This review aspires to provide a comprehensive and detailed overview of current knowledge on corneal therapies involving MSCs and MSC-EVs, the strategies currently under evaluation, and the gaps remaining to be addressed for clinical implementation. From encapsulating MSCs or their EVs into biomaterials to enhance the ocular retention time to loading MSC-EVs with therapeutic drugs, a wide range of ground-breaking strategies are currently contemplated to lead to the safest and most effective treatments. Promising research initiatives also include diverse gene therapies and the targeting of specific cell types through the modification of the EV surface, paving the way for future therapeutic innovations. As one of the most important challenges, MSC-EV large-scale production strategies are extensively investigated and offer a wide array of possibilities to meet the needs of clinical applications.
Collapse
Affiliation(s)
- Manon Jammes
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Abbas Tabasi
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Trung Bach
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland; CURAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
5
|
Narvekar S, Baliga SD, Angadi PV. Dental pulp stem cells as a novel antifibrotic therapy for oral submucous fibrosis: An in vitro study. J Oral Biol Craniofac Res 2025; 15:383-389. [PMID: 40034371 PMCID: PMC11872489 DOI: 10.1016/j.jobcr.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Background Oral submucous fibrosis (OSMF) is a chronic, potentially malignant disorder associated with areca nut consumption. It is characterized by progressive fibrosis, trismus, and a significant risk of malignancy, with limited treatment options primarily offering symptomatic relief. Dental pulp stem cells (DPSCs), a type of mesenchymal stem cells (MSCs), have shown potential for modulating fibrotic conditions through their immunomodulatory and regenerative properties. This study evaluates the antifibrotic potential of DPSCs on OSMF fibroblasts in an in vitro model. Methods DPSCs were isolated from healthy permanent teeth and characterized using flow cytometry for MSC markers (CD73, CD90, CD44, CD105). Fibroblasts were cultured from OSMF biopsy samples and validated through magnetic sorting and morphological analysis. The antifibrotic effects of DPSCs on fibroblasts were evaluated using assays for collagen gel contraction, proliferation, TGF-β1 secretion, and morphological changes. Data were analyzed for statistical significance using appropriate tests. Results The mean collagen gel size decreased from 3.235 mm (95 % CI: 1.65-4.82 mm) in the control group to 1.00 mm (95 % CI: -0.27 - 2.27 mm) in the DPSC-treated group. Fibroblast viability declined significantly over 72 h (p < 0.05). TGF-β1 secretion was markedly lower in DPSC-treated fibroblasts (339.38 pg/mL vs 637.61 pg/mL, p = 0.000393, Cohen's d = 19.15). Conclusion DPSCs exhibit strong antifibrotic properties by inhibiting collagen contraction, suppressing fibroblast proliferation, and reducing TGF-β1 secretion. These findings suggest DPSCs as a promising cell-based therapy for OSMF. Further in vivo studies are warranted for clinical translation. Trial registration number Not applicable.
Collapse
Affiliation(s)
- Snehalata Narvekar
- Department of Oral and Maxillofacial Surgery,KLE V K Institute of Dental Sciences, K LE Academy of Higher Education and Research, Belagavi, India
| | - Shridhar D. Baliga
- Department of Oral and Maxillofacial Surgery, KLE V K Institute of Dental Sciences, KLE Academy of Higher Education and Research, Belagavi, India
| | - Punnya V Angadi
- Department of Oral and Maxillofacial Pathology and Microbiology, KLE V K Institute of Dental Sciences, KLE Academy of Higher Education and Research, Belagavi, India
| |
Collapse
|
6
|
Li X, Jian X, Yan Z, Liu H, Zhang L. Multiple Intra-Articular Injections of Adipose-Derived Mesenchymal Stem Cells for Canine Osteoarthritis Treatment. Cells 2025; 14:323. [PMID: 40072052 PMCID: PMC11899304 DOI: 10.3390/cells14050323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Osteoarthritis (OA) is one of the most common degenerative diseases in dogs and humans, which can lead to articular cartilage deterioration, chronic pain, and decreased quality of life. The anti-inflammatory, anti-fibrotic, analgesic, and cartilage regeneration properties of mesenchymal stem cell (MSC) therapy provide a new direction for the treatment development of OA in the future. Currently, MSC therapy lacks confirmed ideal sources, dosages, formulations, and specific characteristics. In this study, we evaluated the efficacy of multiple canine adipose-derived mesenchymal stem cell (ADSC) injections on anti-inflammation and joint cartilage damage in a canine OA model. Considering animal ethics, we simulated the effects of inflammation and cartilage repair during treatment through a mouse OA model. In the mouse OA model, through the detection of cartilage repair and inflammation-related key factors via histology and molecular biology, it was found that MSC therapy has a certain repair effect on cartilage, but the anti-inflammatory effect is time-dependent. In the canine OA model, we verified the feasibility of multiple injections of ADSCs. Compared with the control group, the cartilage repair effect of the treatment group was obvious, and the inflammatory factors decreased, showing an obvious therapeutic effect. This study demonstrates that multiple intra-articular injections of canine ADSCs could be effective in treating OA symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Lisheng Zhang
- College of Veterinary Medicine/Bio-Medical Center, Huazhong Agricultural University, Wuhan 430070, China; (X.L.); (X.J.); (Z.Y.); (H.L.)
| |
Collapse
|
7
|
Vembuli H, Rajasingh S, Nabholz P, Guenther J, Morrow BR, Taylor MM, Aghazadeh M, Sigamani V, Rajasingh J. Induced mesenchymal stem cells generated from periodontal ligament fibroblast for regenerative therapy. Exp Biol Med (Maywood) 2025; 250:10342. [PMID: 39963344 PMCID: PMC11830513 DOI: 10.3389/ebm.2025.10342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Bone fractures and bone loss represent significant global health challenges, with their incidence rising due to an aging population. Despite autologous bone grafts remain the gold standard for treatment, challenges such as limited bone availability, immune reactions, and the risk of infectious disease transmission have driven the search for alternative cell-based therapies for bone regeneration. Stem cells derived from oral tissues and umbilical cord mesenchymal stem cells (MSCs) have shown potential in both preclinical and clinical studies for bone tissue regeneration. However, their limited differentiation capacity and wound healing abilities necessitate the exploration of alternative cell sources. In this study, we generated induced pluripotent stem cells (iPSCs) using a safe, nonviral and mRNA-based approach from human periodontal ligament fibroblasts (PDLF), an easily accessible cell source. These iPSCs were subsequently differentiated into MSCs, referred to as induced MSCs (iMSCs). The resulting iMSCs were homogeneous, highly proliferative, and possessed anti-inflammatory properties, suggesting their potential as a superior alternative to traditional MSCs for regenerative therapy. These iMSCs demonstrated trilineage differentiation potential, giving rise to osteocytes, chondrocytes, and adipocytes. The iMSC-derived osteocytes (iOSTs) were homogeneous, patient-specific and showed excellent attachment and growth on commercial collagen-based membranes, highlighting their suitability for bone tissue regeneration applications. Given their promising characteristics compared to traditional MSCs, PDLF-derived iMSCs are strong candidates for future clinical studies in bone regeneration and other regenerative dental therapies.
Collapse
Affiliation(s)
- Hemanathan Vembuli
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Patrick Nabholz
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jefferson Guenther
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Brian R. Morrow
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Margaret M. Taylor
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marziyeh Aghazadeh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
8
|
Sheikhi K, Ghaderi S, Firouzi H, Rahimibarghani S, Shabani E, Afkhami H, Yarahmadi A. Recent advances in mesenchymal stem cell therapy for multiple sclerosis: clinical applications and challenges. Front Cell Dev Biol 2025; 13:1517369. [PMID: 39963155 PMCID: PMC11830822 DOI: 10.3389/fcell.2025.1517369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS), is characterized by inflammation, demyelination, and neurodegeneration, leading to diverse clinical manifestations such as fatigue, sensory impairment, and cognitive dysfunction. Current pharmacological treatments primarily target immune modulation but fail to arrest disease progression or entirely reverse CNS damage. Mesenchymal stem cell (MSC) therapy offers a promising alternative, leveraging its immunomodulatory, neuroprotective, and regenerative capabilities. This review provides an in-depth analysis of MSC mechanisms of action, including immune system regulation, promotion of remyelination, and neuroregeneration. It examines preclinical studies and clinical trials evaluating the efficacy, safety, and limitations of MSC therapy in various MS phenotypes. Special attention is given to challenges such as delivery routes, dosing regimens, and integrating MSCs with conventional therapies. By highlighting advancements and ongoing challenges, this review underscores the potential of MSCs to revolutionize MS treatment, paving the way for personalized and combinatory therapeutic approaches.
Collapse
Affiliation(s)
- Kamran Sheikhi
- Kurdistan University of Medical Sciences, Kurdistan, Iran
| | | | - Hassan Firouzi
- Department of Medical Laboratory, Faculty of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Sarvenaz Rahimibarghani
- Department of Physical Medicine and Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Shabani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| |
Collapse
|
9
|
Ozkan S, Isildar B, Koyuturk M. Comparative analysis of the effects of different hypoxia mimetic agents on the secretome contents of conditioned medium obtained from mesenchymal stem/stromal cells cultured in 2 or 3-dimensional cell culture systems. Cytotechnology 2025; 77:11. [PMID: 39654545 PMCID: PMC11625095 DOI: 10.1007/s10616-024-00659-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
Paracrine factors secreted by mesenchymal stem/stromal cells (MSCs) have been demonstrated to have significant therapeutic potential. The secretome profiles of MSCs variate depending on culture conditions. Generally, the effects of a single preconditioning strategy on secretome profiles of MSCs were investigated. However, until now, there has been no study examining the combinatory effects of different preconditioning strategies in a comparative manner. This study aimed to evaluate the secretome contents of conditioned media obtained from human umbilical cord-derived MSCs cultured in 2- or 3-dimensional (D) culture systems preconditioned with deferoxamine (DFS) or dimethyloxalylglycine (DMOG). Immunocytochemical analysis showed that MSCs preconditioned with DFS or DMOG have increased nuclear hypoxia-inducible factor-1α expression. Transmission electron microscopic analysis showed that cells preconditioned with DFS or DMOG have increased autophagic vesicles, which could be attributed to altered energy metabolism under hypoxic conditions. It was revealed that hypoxia-mimetic agents added to the 2D-, or 3D-culture environment raised total protein concentrations per cell along with vascular endothelial growth factor. The concentrations of glial cell-derived neurotrophic factor (GDNF) and nerve growth factor (NGF) were differentially regulated in 2D-, and 3D-culture system, that the secretions of GDNF and NGF per cell were more prominent in 3D- and 2D-culture systems, respectively. These findings indicate that hypoxic conditions alone significantly elevate total protein concentrations, while the contribution of the 3D environment is more modest than initially anticipated. However, concentrations of secreted growth factors may be affected differently depending on the topography of the culture condition and the types of hypoxia mimetic agents.
Collapse
Affiliation(s)
- Serbay Ozkan
- Faculty of Medicine, Histology and Embryology Department, Izmir Katip Çelebi University, Izmir, Turkey
| | - Basak Isildar
- Faculty of Medicine, Histology and Embryology Department, Balıkesir University, Balikesir, Turkey
| | - Meral Koyuturk
- Cerrahpaşa Faculty of Medicine, Histology and Embryology Department, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
10
|
Abbas SEM, Maged G, Wang H, Lotfy A. Mesenchymal Stem/Stromal Cells Microencapsulation for Cell Therapy. Cells 2025; 14:149. [PMID: 39936941 PMCID: PMC11817150 DOI: 10.3390/cells14030149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Cell microencapsulation is one of the most studied strategies to overcome the challenges associated with the implementation of mesenchymal stem/stromal cells (MSCs) in vivo. This approach isolates/shields donor MSCs from the host immune system using a semipermeable membrane that allows for the diffusion of gases, nutrients, and therapeutics, but not host immune cells. As a result, microencapsulated MSCs survive and engraft better after infusion, and they can be delivered specifically to the targeted site. Additionally, microencapsulation enables the co-culture of MSCs with different types of cells in a three-dimensional (3D) environment, allowing for better cellular interaction. Alginate, collagen, and cellulose are the most popular materials, and air jet extrusion, microfluidics, and emulsion are the most used techniques for MSC cell encapsulation in the literature. These materials and techniques differ in the size range of the resultant microcapsules and their compatibility with the applied materials. This review discusses various materials and techniques used for the microencapsulation of MSCs. We also shed light on the recent findings in this field, the advantages and drawbacks of using encapsulated MSCs, and the in vivo translation of the microencapsulated MSCs in cell therapy.
Collapse
Affiliation(s)
| | - Ghada Maged
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21526, Egypt
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Ahmed Lotfy
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
11
|
Paton MCB, Benders M, Blatch-Williams R, Dallimore E, Edwards A, Elwood N, Facer K, Finch-Edmondson M, Garrity N, Gordon A, Hunt RW, Jenkin G, McDonald CA, Moore J, Nold MF, Novak I, Popat H, Salomon C, Sato Y, Tolcos M, Wixey JA, Yawno T, Zhou L, Malhotra A. Updates on neonatal cell and novel therapeutics: Proceedings of the Second Neonatal Cell Therapies Symposium (2024). Pediatr Res 2025:10.1038/s41390-025-03856-x. [PMID: 39815092 DOI: 10.1038/s41390-025-03856-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025]
Abstract
Cell therapies as treatments for neonatal conditions have attracted significant research and parent interest over the last two decades. Mesenchymal stromal cells, umbilical cord blood cells and neural stem cells translate from lab, to preclinical and into clinical trials, with contributions being made from all over the world. Effective and timely translation involves frequent reflection and consultation from research-adjacent fields (i.e. cell therapies for cerebral palsy, adult neurology, companies, and regulatory bodies) as well as meaningful involvement of people with lived experience. Progress to date suggests that aligning outcome and data reporting in later phase clinical trials will support our sector, as well as involving industry partners for streamlined solutions in cell manufacturing, commercialisation and regulatory processes. Importantly, our field can also benefit from resource sharing and research collaboration in novel drug therapies, small molecules and extracellular vesicles as we attempt to bridge preclinical and clinical research. In this review, we present highlights and learnings from the second Neonatal Cell Therapies Symposium (2024), held in Sydney, Australia. IMPACT: Multiple cell therapy candidates have advanced through preclinical and clinical trials in neonatology, showing promising feasibility, safety and efficacy. Effective and timely translation is enabled by collaboration across research-adjacent fields, commercial partnerships, harmonising research outcomes and meaningful involvement of people with lived experience. Progress on the potential utility of cell therapies for neonatal conditions and further translational considerations are discussed in this paper.
Collapse
Affiliation(s)
- Madison C B Paton
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Manon Benders
- Wilhemina Children's Hospital, Neonatology Department, Utrecht Brain Center, University Medical Centre, University Utrecht, Utrecht, The Netherlands
| | - Remy Blatch-Williams
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | | | - Adam Edwards
- Argenica Therapeutics LTD, Nedlands, WA, Australia
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA, Australia
| | - Ngaire Elwood
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- BMDI Cord Blood Bank, Melbourne, VIC, Australia
| | - Kylie Facer
- Parent with Lived Experience, Sydney, Australia
| | - Megan Finch-Edmondson
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Natasha Garrity
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Adrienne Gordon
- Discipline of Obstetrics, Gynaecology, and Neonatology, The University of Sydney, Sydney, NSW, Australia
| | - Rod W Hunt
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Justin Moore
- Department of Neurosurgery, Monash Health, Melbourne, VIC, Australia
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Iona Novak
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Himanshu Popat
- The Children's Hospital at Westmead, Sydney, NSW, Australia
- NHMRC Clinical Trial Centre, University of Sydney, Camperdown, VIC, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Julie A Wixey
- Perinatal Research Centre, University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Tamara Yawno
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia.
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia.
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.
| |
Collapse
|
12
|
Cañas-Arboleda M, Galindo CC, Cruz-Barrera M, Herrera K, Beltrán K, Rodríguez A, Rotter B, Camacho B, Salguero G. Comprehensive analysis of secretome and transcriptome stability of Wharton jelly mesenchymal stromal cells during good manufacturing practice-compliant production. Cytotherapy 2025; 27:107-120. [PMID: 39306795 DOI: 10.1016/j.jcyt.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) hold promise for cell-based therapies due to their ability to stimulate tissue repair and modulate immune responses. Umbilical cord-derived MSCs from Wharton jelly (WJ) offer advantages such as low immunogenicity and potent immune modulatory effects. However, ensuring consistent quality and safety throughout their manufacturing process remains critical. RNA sequencing (RNA-seq) emerges as a crucial tool for assessing genetic stability and expression dynamics in cell-based therapeutic products. METHODS We examined the secretome and transcriptome of WJ-MSC signatures throughout Good Manufacturing Practice (GMP) production, focusing on the performance of total RNA or Massive Analysis of cDNA Ends (MACE) sequencing. RESULTS Through extensive transcriptomic analysis, we demonstrated consistent stability of WJ-MSC expression signatures across different manufacturing stages. Notably, MACE-seq showed improved identification of key expression patterns related to senescence and immunomodulation. CONCLUSIONS These findings highlight the potential of MACE-seq as a quality assessment tool for WJ-MSC-based therapies, ensuring their efficacy and safety in clinical applications. Importantly, MACE-seq demonstrated its value in characterizing WJ-MSC-derived products, offering insights that traditional assays cannot provide.
Collapse
Affiliation(s)
- Mariana Cañas-Arboleda
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia
| | - Cristian Camilo Galindo
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia
| | - Monica Cruz-Barrera
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia
| | - Katherine Herrera
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia
| | - Karl Beltrán
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia
| | | | | | - Bernardo Camacho
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia
| | - Gustavo Salguero
- Advanced Therapy Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá, Colombia.
| |
Collapse
|
13
|
Nasution I, Putra A, Setiawan E. Synergistic Effects of Hypoxia-Preconditioned Mesenchymal Stem Cells Secretome and Alkaline Water in Alleviating Oxidative Stress and Inflammation in Type 2 Diabetic Rats. Mater Sociomed 2025; 37:4-10. [PMID: 40098762 PMCID: PMC11910908 DOI: 10.5455/msm.2024.37.4-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
Background Chronic inflammation and oxidative stress are central to the pathophysiology of Type 2 Diabetes Mellitus (T2DM), contributing to the progression of metabolic dysfunction and related complications. Objective The aim of this study was to explore the therapeutic potential of combining hypoxia-preconditioned mesenchymal stem cell SH-MSC with alkaline water in a T2DM rat model. Methods T2DM was induced in Wistar rats through a high-fat diet (HFD) followed by streptozotocin (STZ) administration. A total of 30 healthy male Wistar rats were randomly assigned to five groups: healthy control, T2DM, T2DM + Metformin, T2DM + SH-MSC, and T2DM + SH-MSC + alkaline water. Results The combination of SH-MSC and alkaline water significantly reduced malondialdehyde (MDA) levels, a key indicator of lipid peroxidation, and suppressed the expression of p65 mRNA, a crucial component of the NF-κB signaling pathway. Notably, the most pronounced reduction in p65 mRNA expression was observed in the group receiving both SH-MSC and alkaline water, suggesting a synergistic effect in mitigating oxidative stress and inflammation. Conclusion These findings highlight the potential of SH-MSC and alkaline water as a novel therapeutic strategy for alleviating T2DM.
Collapse
Affiliation(s)
- Irni Nasution
- Student of Postgraduate Biomedical Science, Universitas Islam Sultan Agung, Semarang, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research (SCCR) Laboratory, Semarang, Indonesia
- Department of Doctoral Biomedical Science, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia
| | - Eko Setiawan
- Department of Postgraduate Biomedical Sciences, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang Indonesia
- Department of Surgery, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia
| |
Collapse
|
14
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
15
|
Nguyen PT, Seo Y, Ahn JS, Oh SJ, Park HJ, Yu JH, Kim SH, Lee Y, Yang JW, Cho J, Kang MJ, Park JH, Kim HS. De novo interleukin-10 production primed by Lactobacillus sakei CVL-001 amplifies the immunomodulatory abilities of mesenchymal stem cells to alleviate colitis. Biomed Pharmacother 2025; 182:117745. [PMID: 39705909 DOI: 10.1016/j.biopha.2024.117745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
Mesenchymal stem cells (MSCs) hold therapeutic promise for treating inflammatory bowel disease (IBD) owing to their immunomodulatory properties. Currently, pre-conditioning strategies with several beneficial agents have been applied to enhance the efficacy of MSCs in treating IBDs. Probiotics are increasingly acknowledged as supplemental therapy for IBD; however, their potential benefits in MSCs-based therapy remain largely unexplored. In this study, we hypothesized that pretreating MSCs with Lactobacillus sakei CVL-001 (L. sakei CVL-001), a representative probiotic strain, could improve their therapeutic effectiveness for IBD. In line with this hypothesis, we noted that pretreatment with L. sakei CVL-001 significantly induced IL-10 secretion in MSCs via the activation of the STAT3 signaling pathway. These primed MSCs reduced pro-inflammatory cytokine production in LPS/IFN-γ-treated macrophages and promoted an M2 phenotype, associated with immunoregulation and tissue repair, in undifferentiated macrophages. In addition, their conditioned media significantly reduced the proliferation capacity of Jurkat T cells and splenocytes, while the neutralization of IL-10 reversed these phenomena. Furthermore, MSCs treated with L. sakei CVL-001 mitigated inflammatory responses and promoted epithelial regeneration, leading to accelerated recovery from disease symptoms and improved survival rates compared to naive MSCs in a DSS-induced colitis mouse model. In conclusion, our findings suggest that probiotics, such as L. sakei CVL-001, can improve the therapeutic efficacy of MSCs for treating IBD.
Collapse
Affiliation(s)
- Phuong Thao Nguyen
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Su-Jeong Oh
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jeong Hyun Yu
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seong Hui Kim
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yunji Lee
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji Won Yang
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jaejin Cho
- Department of Dental Regenerative Biotechnology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea; NODCURE, Inc, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea.
| | - Hyung-Sik Kim
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
16
|
Erol Bozkurt A, Sel FA, Suleymanoğlu M, Demirayak G, Kuruca DS, Oğuz FS. The Cytokine Levels of Cord Blood- and Wharton's Jelly-Derived Mesenchymal Stem Cells from Early to Late Passages. Cell Biochem Biophys 2024; 82:3345-3350. [PMID: 39018006 DOI: 10.1007/s12013-024-01416-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2024] [Indexed: 07/18/2024]
Abstract
Mesenchymal stem cells (MSCs) are promising for clinical studies owing to their self-renewal, multipotency, trophic, and immunomodulatory properties. This study aimed to investigate the cytokine levels of human umbilical cord blood (CB) and Wharton's Jelly-(WJ) derived MSCs relevant to immune modulation on different passage levels in vitro. Umbilical CB MSCs were isolated using the ficoll-paque gradient method, and WJ-MSCs were isolated by the explant method. After isolation, the MSCs were characterized using flow cytometry. The supernatant cytokine levels (interferon-gamma (IFN-γ), interleukin 4 (IL-4), interleukin 17 (IL-17)) of MSCs at each passage were evaluated using the ELISA assay. MSCs exhibited different cytokine levels with each passage number. In WJ-MSC culture supernatants, IL-17 levels significantly increased at P4 and P5 compared to the first passage (p < 0.005), while the other passages showed a decrease. IFN-γ levels increased at passage P1 and P4 and decreased at other passages (p < 0.005). IL-4 levels significantly increased only at passage P3 (p < 0.005). In CB-MSC culture supernatants, IL-17 and IL-4 cytokines decreased compared to P0, while IFN-γ cytokine increased from P0 (p < 0.005). The changing ratio of cytokine levelsfor both CB-MSCs and WJ-MSCs were similarly maintained from early to late passages. More research is needed to understand the immunomodulatory functions of MSCs.
Collapse
Affiliation(s)
- Ayşe Erol Bozkurt
- Istanbul University, Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul, Türkiye.
| | - Figen Abatay Sel
- Istanbul University, Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul, Türkiye
| | - Mediha Suleymanoğlu
- Istanbul University, Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul, Türkiye
| | - Gökhan Demirayak
- University of Health Sciences, Bakırköy Sadi Konuk Education and Research Hospital, Department of Gynecologic Oncology, Istanbul, Türkiye
| | - Dürdane Serap Kuruca
- Istanbul University, Istanbul Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Fatma Savran Oğuz
- Istanbul University, Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul, Türkiye
| |
Collapse
|
17
|
Wu KC, Chang YH, Ding DC, Lin SZ. Mesenchymal Stromal Cells for Aging Cartilage Regeneration: A Review. Int J Mol Sci 2024; 25:12911. [PMID: 39684619 PMCID: PMC11641625 DOI: 10.3390/ijms252312911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cartilage degeneration is a key feature of aging and osteoarthritis, characterized by the progressive deterioration of joint function, pain, and limited mobility. Current treatments focus on symptom relief, not cartilage regeneration. Mesenchymal stromal cells (MSCs) offer a promising therapeutic option due to their capability to differentiate into chondrocytes, modulate inflammation, and promote tissue regeneration. This review explores the potential of MSCs for cartilage regeneration, examining their biological properties, action mechanisms, and applications in preclinical and clinical settings. MSCs derived from bone marrow, adipose tissue, and other sources can self-renew and differentiate into multiple cell types. In aging cartilage, they aid in tissue regeneration by secreting growth factors and cytokines that enhance repair and modulate immune responses. Recent preclinical studies show that MSCs can restore cartilage integrity, reduce inflammation, and improve joint function, although clinical translation remains challenging due to limitations such as cell viability, scalability, and regulatory concerns. Advancements in MSC delivery, including scaffold-based approaches and engineered exosomes, may improve therapeutic effectiveness. Potential risks, such as tumorigenicity and immune rejection, are also discussed, emphasizing the need for optimized treatment protocols and large-scale clinical trials to develop effective, minimally invasive therapies for cartilage regeneration.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
18
|
Yordanova A, Ivanova M, Tumangelova-Yuzeir K, Angelov A, Kyurkchiev S, Belemezova K, Kurteva E, Kyurkchiev D, Ivanova-Todorova E. Umbilical Cord Mesenchymal Stem Cell Secretome: A Potential Regulator of B Cells in Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:12515. [PMID: 39684227 DOI: 10.3390/ijms252312515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Autoimmune diseases represent a severe personal and healthcare problem that seeks novel therapeutic solutions. Mesenchymal stem cells (MSCs) are multipotent cells with interesting cell biology and promising therapeutic potential. The immunoregulatory effects of secretory factors produced by umbilical cord mesenchymal stem cells (UC-MSCs) were assessed on B lymphocytes from 17 patients with systemic lupus erythematosus (SLE), as defined by the 2019 European Alliance of Associations for Rheumatology (EULAR)/American College of Rheumatology (ACR) classification criteria for SLE, and 10 healthy volunteers (HVs). Peripheral blood mononuclear cells (PBMCs) from patients and HVs were cultured in a UC-MSC-conditioned medium (UC-MSCcm) and a control medium. Flow cytometry was used to detect the surface expression of CD80, CD86, BR3, CD40, PD-1, and HLA-DR on CD19+ B cells and assess the percentage of B cells in early and late apoptosis. An enzyme-linked immunosorbent assay (ELISA) quantified the production of BAFF, IDO, and PGE2 in PBMCs and UC-MSCs. Under UC-MSCcm influence, the percentage and mean fluorescence intensity (MFI) of CD19+BR3+ cells were reduced in both SLE patients and HVs. Regarding the effects of the MSC secretome on B cells in lupus patients, we observed a decrease in CD40 MFI and a reduced percentage of CD19+PD-1+ and CD19+HLA-DR+ cells. In contrast, in the B cells of healthy participants, we found an increased percentage of CD19+CD80+ cells and decreased CD80 MFI, along with a decrease in CD40 MFI and the percentage of CD19+PD-1+ cells. The UC-MSCcm had a minimal effect on B-cell apoptosis. The incubation of patients' PBMCs with the UC-MSCcm increased PGE2 levels compared to the control medium. This study provides new insights into the impact of the MSC secretome on the key molecules involved in B-cell activation and antigen presentation and survival, potentially guiding the development of future SLE treatments.
Collapse
Affiliation(s)
- Adelina Yordanova
- University Hospital St. Ivan Rilski, Laboratory of Clinical Immunology, Department of Clinical Immunology, Medical Faculty, Medical University of Sofia, 15 Akademik Iv. E. Geshov Blvd., 1431 Sofia, Bulgaria
| | - Mariana Ivanova
- University Hospital St. Ivan Rilski, Clinic of Rheumatology, Department of Rheumatology, Medical Faculty, Medical University of Sofia, 13 Urvich St., 1612 Sofia, Bulgaria
| | - Kalina Tumangelova-Yuzeir
- University Hospital St. Ivan Rilski, Laboratory of Clinical Immunology, Department of Clinical Immunology, Medical Faculty, Medical University of Sofia, 15 Akademik Iv. E. Geshov Blvd., 1431 Sofia, Bulgaria
| | - Alexander Angelov
- University Hospital St. Ivan Rilski, Clinic of Rheumatology, Department of Rheumatology, Medical Faculty, Medical University of Sofia, 13 Urvich St., 1612 Sofia, Bulgaria
| | | | | | - Ekaterina Kurteva
- University Hospital St. Ivan Rilski, Laboratory of Clinical Immunology, Department of Clinical Immunology, Medical Faculty, Medical University of Sofia, 15 Akademik Iv. E. Geshov Blvd., 1431 Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- University Hospital St. Ivan Rilski, Laboratory of Clinical Immunology, Department of Clinical Immunology, Medical Faculty, Medical University of Sofia, 15 Akademik Iv. E. Geshov Blvd., 1431 Sofia, Bulgaria
| | - Ekaterina Ivanova-Todorova
- University Hospital St. Ivan Rilski, Laboratory of Clinical Immunology, Department of Clinical Immunology, Medical Faculty, Medical University of Sofia, 15 Akademik Iv. E. Geshov Blvd., 1431 Sofia, Bulgaria
| |
Collapse
|
19
|
Dairov A, Sekenova A, Alimbek S, Nurkina A, Shakhatbayev M, Kumasheva V, Kuanysh S, Adish Z, Issabekova A, Ogay V. Psoriasis: The Versatility of Mesenchymal Stem Cell and Exosome Therapies. Biomolecules 2024; 14:1351. [PMID: 39595528 PMCID: PMC11591958 DOI: 10.3390/biom14111351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are multilineage differentiating stromal cells with extensive immunomodulatory and anti-inflammatory properties. MSC-based therapy is widely used in the treatment of various pathologies, including bone and cartilage diseases, cardiac ischemia, diabetes, and neurological disorders. Along with MSCs, it is promising to study the therapeutic properties of exosomes derived from MSCs (MSC-Exo). A number of studies report that the therapeutic properties of MSC-Exo are superior to those of MSCs. In particular, MSC-Exo are used for tissue regeneration in various diseases, such as healing of skin wounds, cancer, coronary heart disease, lung injury, liver fibrosis, and neurological, autoimmune, and inflammatory diseases. In this regard, it is not surprising that the scientific community is interested in studying the therapeutic properties of MSCs and MSC-Exo in the treatment of psoriasis. This review summarizes the recent advancements from preclinical and clinical studies of MSCs and MSC-Exo in the treatment of psoriasis, and it also discusses their mechanisms of therapeutic action involved in the treatment of this disease.
Collapse
Affiliation(s)
- Aidar Dairov
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
- Department of General Biology and Genomics, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Aliya Sekenova
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
| | - Symbat Alimbek
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
| | - Assiya Nurkina
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
| | - Miras Shakhatbayev
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
| | - Venera Kumasheva
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
| | - Sandugash Kuanysh
- Obstetrics and Gynecology, Astana Medical University, Astana 010000, Kazakhstan
| | - Zhansaya Adish
- Laboratory of Immunochemistry and Immunobiotechnology, National Center for Biotechnology, Astana 010000, Kazakhstan;
- Department of Natural Sciences, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Assel Issabekova
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
| | - Vyacheslav Ogay
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan or (A.D.); (A.S.); (S.A.); (A.N.); (M.S.); (V.K.); (V.O.)
- Department of General Biology and Genomics, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| |
Collapse
|
20
|
Ma D, Feng Y, Lin X. Immune and non-immune mediators in the fibrosis pathogenesis of salivary gland in Sjögren's syndrome. Front Immunol 2024; 15:1421436. [PMID: 39469708 PMCID: PMC11513355 DOI: 10.3389/fimmu.2024.1421436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Sjögren's syndrome (SS) or Sjögren's disease (SjD) is a systemic autoimmune disease clinically manifested as sicca symptoms. This disease primarily impacts the functionality of exocrine glands, specifically the lacrimal and salivary glands (SG). SG fibrosis, an irreversible morphological change, is a severe consequence that occurs in the later stages of the disease due to sustained inflammation. However, the mechanism underlying SG fibrosis in SS remains under-investigated. Glandular fibrosis may arise from chronic sialadenitis, in which the interactions between infiltrating lymphocytes and epithelial cells potentially contributes to fibrotic pathogenesis. Thus, both immune and non-immune cells are closely involved in this process, while their interplays are not fully understood. The molecular mechanism of tissue fibrosis is partly associated with an imbalance of immune responses, in which the transforming growth factor-beta (TGF-β)-dependent epithelial-mesenchymal transition (EMT) and extracellular matrix remodeling are recently investigated. In addition, viral infection has been implicated in the pathogenesis of SS. Viral-specific innate immune response could exacerbate the autoimmune progression, resulting in overt inflammation in SG. Notably, post-COVID patients exhibit typical SS symptoms and severe inflammatory sialadenitis, which are positively correlated with SG damage. In this review, we discuss the immune and non-immune risk factors in SG fibrosis and summarize the evidence to understand the mechanisms upon autoimmune progression in SS.
Collapse
Affiliation(s)
- Danbao Ma
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Xiang Lin
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Chinese Medicine, the University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China
| |
Collapse
|
21
|
Galhom RA, Ali SNS, El-Fark MMO, Ali MHM, Hussein HH. Assessment of therapeutic efficacy of adipose tissue-derived mesenchymal stem cells administration in hyperlipidemia-induced aortic atherosclerosis in adult male albino rats. Tissue Cell 2024; 90:102498. [PMID: 39079452 DOI: 10.1016/j.tice.2024.102498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024]
Abstract
Atherosclerosis (AS) is a common disease seriously detrimental to human health. AS is a chronic progressive disease related to inflammatory reactions. The present study aimed to characterize and evaluate the effects of adipose tissue stem cells (ADSCs) in high-fat diet-induced atherosclerosis in a rat model. The present study comprises thirty-six rats and they were divided into three groups: the control group, the high-fat diet (HFD) group; which received a high-fat diet, and the high-fat diet + stem cells (HFD+SC) group; which was fed with a high-fat diet along with the administration of intravenous ADSCs. Food was given to the animals for 20 weeks to establish dyslipidemia models. After 20 weeks, animals were sacrificed by cervical dislocation; blood was collected to measure total cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL); aortae were collected to detect morphologic changes. Rats of the HFD group showed a significant increase in body weight (B.Wt), altered lipid profile increased expression of inducible nitric oxide synthase (iNOS), and decreased expression of endothelial nitric oxide synthase (eNOS). However, in HFD+SC there was a significant decrease in body weight gain and an improvement in lipid profile. Histopathological and ultrastructural variations observed in the aorta of the HFD group when treated with ADSCs showed preserved normal histological architecture and reduced atherosclerosis compared with the HFD group. This was evidenced by laboratory, histological, immunohistochemical, and morphometric studies. Thus, ADSCs reduced TC, TG, and LDL, reduced the expression of iNOS, and increased the expression of eNOS. The high-fat diet was likely to cause damage to the wall of blood vessels. Systemically transplanted ADSCs could home to the aorta, and further protect the aorta from HFD-induced damage.
Collapse
Affiliation(s)
- Rania A Galhom
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Department of Human Anatomy and Embryology, Faculty of Medicine, Badr University in Cairo (BUC), Egypt.
| | - Saleh Nasser Saleh Ali
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Department of Human Anatomy and Embryology, Faculty of Medicine and Health Sciences, Thamar University, Thamar, Yemen.
| | - Magdy Mohamed Omar El-Fark
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mona Hassan Mohammed Ali
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Hoda Hassan Hussein
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
22
|
Işıldar B, Özkan S, Koyutürk M. Preconditioning of Human Umbilical Cord Mesenchymal Stem Cells with a Histone Deacetylase Inhibitor: Valproic Acid. Balkan Med J 2024; 41:369-376. [PMID: 39239940 PMCID: PMC11588919 DOI: 10.4274/balkanmedj.galenos.2024.2024-6-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) play a key role in regenerative medicine due to their capacity to differentiate into multiple cell lines, regulate the immune system, and exert paracrine effects. The therapeutic impact of MSCs is primarily mediated through their secretome. The secretory and therapeutic potential of MSCs can be improved through preconditioning, which entails the application of hypoxic environments, 3-dimensional cell cultures, and pharmacological agents. Valproic acid (VPA) is a histone deacetylase inhibitor that is employed in medical practice for treating epilepsy and bipolar disorder. Hence, preconditioning MSCs with VPA is expected to induce histone acetylation, enhance gene expression, and beneficially modify the cells' secretomes. Aims To assess the effectiveness of VPA in enhancing and regulating the therapeutic potential of cells as well as its impact on MSC secretome profiles and ultrastructural morphologies. Study Design Expiremental study. Methods Human umbilical cord MSCs were preconditioned with 2 mM VPA for 24 and 48 hours; untreated MSCs served as controls. The secretome secreted by the cells was assessed for its total protein content. Subsequently, interferon-gamma (IFN-γ), interleukin-17 (IL-17), IL-10, vascular endothelial growth factor, nerve growth factor (NGF), glial cell line-derived neurotrophic factor, and brain-derived neurotrophic factor (BDNF) levels in the secretome were analyzed using the ELISA method. The ultrastructural properties of the cells were studied under transmission electron microscopy. Results Ultrastructural examinations revealed that the chromatin content of VPA-treated cells was reduced. VPA-preconditioned cells exhibited a higher density of rough endoplasmic reticulum, autophagic vesicles, and myelin figures on cytoplasmic structure analysis, which was indicative of increased secretion. Protein secretion was elevated in those cells, with notable increases in NGF and BDNF levels. Furthermore, the cytoskeletal rearrangement and elevated autophagic activity observed in the 48-hour preconditioned cells could indicate the initiation of neuronal differentiation. IL-10, IL-17, and IFN-γ were not detected in the secretome. Conclusion This study indicate that preconditioning with VPA enhances MSC activity and subsequently modifies the secretome content.
Collapse
Affiliation(s)
- Başak Işıldar
- Department of Histology and Embryology İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Türkiye
- Department of Histology and Embryology Balıkesir University Faculty of Medicine, Balıkesir, Türkiye
| | - Serbay Özkan
- Department of Histology and Embryology İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Türkiye
- Department of Histology and Embryology İzmir Katip Çelebi University Faculty of Medicine, İzmir, Türkiye
| | - Meral Koyutürk
- Department of Histology and Embryology İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Türkiye
| |
Collapse
|
23
|
Lin S, Chang Y, Lee W, Chiang C, Liu S, Lee H, Jeng L, Shyu W. Role of STAT3-FOXO3 Signaling in the Modulation of Neuroplasticity by PD-L1-HGF-Decorated Mesenchymal Stem Cell-Derived Exosomes in a Murine Stroke Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404882. [PMID: 39049677 PMCID: PMC11423231 DOI: 10.1002/advs.202404882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/28/2024] [Indexed: 07/27/2024]
Abstract
The limited therapeutic strategies available for stroke leave many patients disabled for life. This study assessed the potential of programmed death-ligand 1 (PD-L1) and hepatocyte growth factor (HGF)-engineered mesenchymal stem cell-derived exosomes (EXO-PD-L1-HGF) in enhancing neurological recovery post-stroke. EXO-PD-L1-HGF, which efficiently endocytosed into target cells, significantly diminishes the H2O2-induced neurotoxicity and increased the antiapoptotic proteins in vitro. EXO-PD-L1-HGF attenuates inflammation by inhibiting T-cell proliferation and increasing the number of CD8+CD122+IL-10+ regulatory T cells. Intravenous injection of EXO-PD-L1-HGF could target stromal cell-derived factor-1α (SDF-1α+) cells over the peri-infarcted area of the ischemic brain through CXCR4 upregulation and accumulation in neuroglial cells post-stroke. EXO-PD-L1-HGF facilitates endogenous nestin+ neural progenitor cell (NPC)-induced neurogenesis via STAT3-FOXO3 signaling cascade, which plays a pivotal role in cell survival and neuroprotection, thereby mitigating infarct size and enhancing neurological recovery in a murine stroke model. Moreover, increasing populations of the immune-regulatory CD19+IL-10+ and CD8+CD122+IL-10+ cells, together with reducing populations of proinflammatory cells, created an anti-inflammatory microenvironment in the ischemic brain. Thus, innovative approaches employing EXO-PD-L1-HGF intervention, which targets SDF-1α+ expression, modulates the immune system, and enhances the activation of resident nestin+ NPCs, might significantly alter the brain microenvironment and create a niche conducive to inducing neuroplastic regeneration post-stroke.
Collapse
Affiliation(s)
- Syuan‐Ling Lin
- Translational Medicine Research Center and Department of NeurologyChina Medical University HospitalTaichung404Taiwan
| | - Yi‐Wen Chang
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
- Department of Medical ResearchNational Taiwan University HospitalTaipei100Taiwan
| | - Wei Lee
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
| | - Chih‐Sheng Chiang
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
- Graduate Institute of Biomedical Sciences and New Drug Development CenterChina Medical UniversityTaichung404Taiwan
| | - Shih‐Ping Liu
- Translational Medicine Research Center and Department of NeurologyChina Medical University HospitalTaichung404Taiwan
- Graduate Institute of Biomedical Sciences and New Drug Development CenterChina Medical UniversityTaichung404Taiwan
| | - Hsu‐Tung Lee
- Graduate Institute of Medical SciencesNational Defense Medical CenterTaipei114Taiwan
- Department of Post‐Baccalaureate Medicine, College of MedicineNational Chung Hsing UniversityTaichung402Taiwan
- Division of neurosurgical Oncology Neurological InstituteTaichung Veterans General HospitalTaichung407Taiwan
| | - Long‐Bin Jeng
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
- Organ Transplantation CenterChina Medical University HospitalTaichung404Taiwan
| | - Woei‐Cherng Shyu
- Translational Medicine Research Center and Department of NeurologyChina Medical University HospitalTaichung404Taiwan
- Graduate Institute of Biomedical Sciences and New Drug Development CenterChina Medical UniversityTaichung404Taiwan
- Department of Occupational TherapyAsia UniversityTaichung413Taiwan
| |
Collapse
|
24
|
Kaundal U, Rakha A. Differential effects of TLR3 and TLR4 activation on MSC-mediated immune regulation. Biochem Biophys Rep 2024; 39:101809. [PMID: 39228386 PMCID: PMC11369377 DOI: 10.1016/j.bbrep.2024.101809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) have evolved as an invaluable therapeutic cell type due to their broad therapeutic properties. Bone marrow-derived MSCs are currently being applied in numerous clinical trials, and the initial results have been encouraging. However, heterogeneous responsiveness amongst patients is also being experienced; therefore, the efficacy of MSCs in vivo is still debatable. Host microenvironment plays an essential role in determining the fate of MSCs in vivo. Recent studies have indicated the role of toll-like receptors (TLR) in modulating the biological properties of MSCs. TLRs are expressed by MSCs, and activation of TLR3 and TLR4 can alter the functionality of MSCs. While MSCs can suppress the effector and memory T cell function by promoting regulatory T cells, the effect of TLR activation on MSC-mediated immune cell induction is still not well understood. This study was performed to understand the TLR licensing of MSCs and its impact on MSC-mediated immunomodulation. We found that TLR3 mediated activation of MSCs (TLR3-MSCs) increased the expression of G-CSF & IL-10 while TLR4-mediated activation of MSCs led to an increase in CXCL-1, CXCL-10, and CXCL-12. To study the immunological aspect, an in vitro co-culture model was established-to imitate the brief in vivo interaction of MSCs and immune cells. We found that TLR3-MSCs led to increase in CD4 and CD8 naive T (TNAI) cells and vice versa for effector (TEFF) and memory T (TMEM) cells, while TLR4-MSCs did not show any effect. Moreover, only TLR3-MSCs led to a non-significant increase in the regulatory T cells (TREGS) and Double negative regulatory cells. No change in B cell profile was evident while TLR3-MSCs depicted an increasing trend in regulatory B cells which was not statistically significant. TLR3 MSCs also inhibited the T cell proliferation in our setup. Our data indicate that TLR3 priming may regulate the function of MSCs through immunomodulation. Understanding the role of TLRs and other microenvironmental factors causing subdued responses of MSCs in vivo would allow the uninhibited use of MSCs for many diseased conditions.
Collapse
Affiliation(s)
- Urvashi Kaundal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Sector-12, Chandigarh, 160012, India
- Scleroderma Genomics and Health Disparities Unit, NIAMS, NIH, Bethesda, USA
| | - Aruna Rakha
- Scleroderma Genomics and Health Disparities Unit, NIAMS, NIH, Bethesda, USA
| |
Collapse
|
25
|
Radan M, Abol Nejadian F, Bayati V, Hemmati AA, Hoseinynejad K, Mard SA. N-acetyl cysteine augments adipose tissue-derived stem cell efficacy on inflammatory markers and regulatory T cell system balance in an allergic asthma model. J Asthma 2024; 61:1029-1041. [PMID: 38376812 DOI: 10.1080/02770903.2024.2321296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Allergic asthma is a destructive inflammatory process in the respiratory system. The anti-inflammatory and antioxidant effects of N-acetylcysteine (NAC) have been reported in patients with obstructive pulmonary disease. On the other hand, several studies have shown the modulatory effects of mesenchymal stem cells on the immune system and inflammatory responses. Accordingly, the purpose of the current study was to evaluate the effect of administration of adipose tissue-derived stem cells (ADSCs) plus NAC on regulatory T cell system balance in an allergic asthma model. METHODS Eighty Sprague- Dawley rats were randomly divided into the following groups: Control, Plasmalite, Allergic asthma, Allergic asthma + ADSCs, NAC, Allergic asthma + NAC, Allergic asthma + ADSCs + NAC and Allergic asthma + Prednisolone. at the end of the experiment, arterial blood gas analysis, inflammatory cell counts in bronchoalveolar lavage fluid (BALF), inflammatory cytokine concentration, total IgE and specific OVA-IgE levels, gene expression levels of CD4+-T cell subsets, pulmonary indicators, edema, and lung histopathology were evaluated in all groups. RESULTS Administration of NAC plus ADSCs demonstrated a significant decrease in total WBC and eosinophil counts, which was in line with remarkable decrease in IL-17 and TNF-α concentrations and increases in IL-10 level compared with other treated groups. NAC plus ADSC treatment showed significant increases in Treg gene expression, although Th17 and Th2 expression significantly decreased compared with that in prednisolone- treated rats. CONCLUSION The results of the present study documented that the administration of ADSCs plus NAC has an inhibitory effect on the inflammation caused by allergic asthma in a rat model. The improvement of inflammatory indexes was significantly higher than that with prednisolone treatment.
Collapse
Affiliation(s)
- Maryam Radan
- Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farhad Abol Nejadian
- Department of Pediatrics, Abuzar Children's Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Asghar Hemmati
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Khojasteh Hoseinynejad
- Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
26
|
Priyadarshani P, Van Grouw A, Liversage AR, Rui K, Nikitina A, Tehrani KF, Aggarwal B, Stice SL, Sinha S, Kemp ML, Fernández FM, Mortensen LJ. Investigation of MSC potency metrics via integration of imaging modalities with lipidomic characterization. Cell Rep 2024; 43:114579. [PMID: 39153198 DOI: 10.1016/j.celrep.2024.114579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/17/2024] [Accepted: 07/18/2024] [Indexed: 08/19/2024] Open
Abstract
Mesenchymal stem/stromal cell (MSC) therapies have had limited success so far in clinical trials due in part to heterogeneity in immune-responsive phenotypes. Therefore, techniques to characterize these properties of MSCs are needed during biomanufacturing. Imaging cell shape, or morphology, has been found to be associated with MSC immune responsivity-but a direct relationship between single-cell morphology and function has not been established. We used label-free differential phase contrast imaging and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to evaluate single-cell morphology and explore relationships with lipid metabolic immune response. In interferon gamma (IFN-γ)-stimulated MSCs, we found higher lipid abundances from the ceramide-1-phosphate (C1P), phosphatidylcholine (PC), LysoPC, and triglyceride (TAG) families that are involved in cell immune function. Furthermore, we identified differences in lipid signatures in morphologically defined MSC subpopulations. The use of single-cell optical imaging coupled with single-cell spatial lipidomics could assist in optimizing the MSC production process and improve mechanistic understanding of manufacturing process effects on MSC immune activity and heterogeneity.
Collapse
Affiliation(s)
- Priyanka Priyadarshani
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Alexandria Van Grouw
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Adrian Ross Liversage
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Kejie Rui
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Arina Nikitina
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kayvan Forouhesh Tehrani
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL 61820, USA
| | - Bhavay Aggarwal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Steven L Stice
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Saurabh Sinha
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Luke J Mortensen
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
27
|
Chen YH, Hung YP, Chen CY, Chen YT, Tsai TC, Yang JJ, Wu CC. ELIXCYTE ®, an Allogenic Adipose-Derived Stem Cell Product, Mitigates Osteoarthritis by Reducing Inflammation and Preventing Cartilage Degradation In Vitro. Curr Issues Mol Biol 2024; 46:8395-8406. [PMID: 39194712 DOI: 10.3390/cimb46080495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) comprise a promising therapy for osteoarthritis (OA). The therapeutic potential of ELIXCYTE®, an allogeneic human ADSC (hADSC) product, was demonstrated in a phase I/II OA clinical trial. However, the exact mechanism underlying such effects is not clear. Moreover, studies suggest that interleukin-11 (IL-11) has anti-inflammatory, tissue-regenerative, and immune-regulatory functions. Our aim was to unravel the mechanism associated with the therapeutic effects of ELIXCYTE® on OA and its relationship with IL-11. We cocultured ELIXCYTE® with normal human articular chondrocytes (NHACs) in synovial fluid obtained from individuals with OA (OA-SF) to investigate its effect on chondrocyte matrix synthesis and degradation and inflammation by assessing gene expression and cytokine levels. NHACs exposed to OA-SF exhibited increased MMP13 expression. However, coculturing ELIXCYTE® with chondrocytes in OA-SF reduced MMP13 expression in chondrocytes and downregulated PTGS2 and FGF2 expression in ELIXCYTE®. ELIXCYTE® treatment elevated anti-inflammatory cytokine (IL-1RA, IL-10, and IL-13) levels, and the reduction in MMP13 was positively correlated with IL-11 concentrations in OA-SF. These findings indicate that IL-11 in OA-SF might serve as a predictive biomarker for the ELIXCYTE® treatment response in OA, emphasizing the therapeutic potential of ELIXCYTE® to mitigate OA progression and provide insights into its immunomodulatory effects.
Collapse
Affiliation(s)
- Yu-Hsiu Chen
- Rheumatology/Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Yi-Pei Hung
- UnicoCell Biomed Co., Ltd., Taipei 11494, Taiwan
| | | | - Yi-Ting Chen
- UnicoCell Biomed Co., Ltd., Taipei 11494, Taiwan
| | | | - Jui-Jung Yang
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Chia-Chun Wu
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| |
Collapse
|
28
|
Wei D, Liang X, Huang M, Wang C, Ye Z, Zhang T, Zhang J. Targeting histone deacetylase 1 (HDAC1) in the bone marrow stromal cells revers imatinib resistance by modulating IL-6 in Ph + acute lymphoblastic leukemia. Ann Hematol 2024; 103:3015-3027. [PMID: 38847852 DOI: 10.1007/s00277-024-05830-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/31/2024] [Indexed: 07/28/2024]
Abstract
Bone marrow stromal cells (BMSCs) can promote the growth of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Histone deacetylases (HDACs) play essential roles in the proliferation and apoptosis resistance of Ph + ALL cells. In our previous study, inhibiting histone deacetylase 1 (HDAC1) decreases the proliferation of Ph + ALL cells. However, little is known regarding how HDAC1 in BMSCs of Ph + ALL patients affects the imatinib (IM) resistance. Therefore, the present work examined the roles of HDAC1 in BMSCs. Overexpression of HDAC1 was found in BMSCs of Ph + ALL patients with IM resistance. In addition, the Ph + ALL cell line SUP-B15 was co-cultured with BMSCs after lentivirus transfection for regulating HDAC1 expression. Knockdown of HDAC1 within BMSCs elevated the IM-mediated SUP-B15 cell apoptosis, while increasing HDAC1 expression had an opposite effect. IL-6 in BMSCs, which is an important factor for the microenvironment-associated chemoresistance, showed evident up-regulation in HDAC1-upregulated BMSCs and down-regulation in HDAC1-downregulated BMSCs. While recombinant IL-6 (rIL-6) can reversed the sensitivity of SUP-B15 cells to IM induced by downregulating HDAC1 expression in BMSCs. HDAC1 showed positive regulation on IL-6 transcription and secretion. Moreover, IL-6 secretion induced by HDAC1 in BMSCs might enhance IM resistance in Ph + ALL cells. With regard to the underlying molecular mechanism, NF-κB, an important signal responsible for IL-6 transcription in BMSCs, mediated the HDAC1-regulated IL-6 expression. Collectively, this study facilitated to develop HDAC1 inhibitors based not only the corresponding direct anti-Ph + ALL activity but also the regulation of bone marrow microenvironment.
Collapse
Affiliation(s)
- Danna Wei
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Xiaoling Liang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Meiling Huang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Caili Wang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Zhangmin Ye
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Jingrong Zhang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China.
| |
Collapse
|
29
|
Ahmed LA, Al-Massri KF. Exploring the Role of Mesenchymal Stem Cell-Derived Exosomes in Diabetic and Chemotherapy-Induced Peripheral Neuropathy. Mol Neurobiol 2024; 61:5916-5927. [PMID: 38252384 PMCID: PMC11249772 DOI: 10.1007/s12035-024-03916-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 12/31/2023] [Indexed: 01/23/2024]
Abstract
Diabetic and chemotherapy-induced peripheral neuropathies are known for long-term complications that are associated with uncontrolled hyperglycemia and cancer treatment, respectively. Peripheral neuropathy often requires long-term therapy and could persist after treatment provoking detrimental effects on the patient's quality of life. Despite continuous drug discoveries, development of efficient therapies is still needed for the significant management of diabetic and chemotherapy-induced peripheral neuropathy. Exosomes are nanosized extracellular vesicles that show great promise recently in tissue regeneration and injury repair compared to their parent stem cells. Herein, we provided a summary for the use of mesenchymal stem cell-derived exosomes in diabetic and chemotherapy-induced peripheral neuropathy in addition to recent advancements and ways proposed for the enhancement of their efficacy in these diseases.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt.
| | - Khaled F Al-Massri
- Department of Pharmacy and Biotechnology, Faculty of Medicine and Health Sciences, University of Palestine, Gaza, Palestine
| |
Collapse
|
30
|
Su W, Yin Y, Zhao J, Hu R, Zhang H, Hu J, Ren R, Zhang Y, Wang A, Lyu Z, Mu Y, Cheng Y. Exosomes derived from umbilical cord-derived mesenchymal stem cells exposed to diabetic microenvironment enhance M2 macrophage polarization and protect against diabetic nephropathy. FASEB J 2024; 38:e23798. [PMID: 38989582 DOI: 10.1096/fj.202400359r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/07/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
The role of mesenchymal-stem-cell-derived exosomes (MSCs-Exo) in the regulation of macrophage polarization has been recognized in several diseases. There is emerging evidence that MSCs-Exo partially prevent the progression of diabetic nephropathy (DN). This study aimed to investigate whether exosomes secreted by MSCs pre-treated with a diabetic environment (Exo-pre) have a more pronounced protective effect against DN by regulating the balance of macrophages. Exo-pre and Exo-Con were isolated from the culture medium of UC-MSCs pre-treated with a diabetic mimic environment and natural UC-MSCs, respectively. Exo-pre and Exo-Con were injected into the tail veins of db/db mice three times a week for 6 weeks. Serum creatinine and serum urea nitrogen levels, the urinary protein/creatinine ratio, and histological staining were used to determine renal function and morphology. Macrophage phenotypes were analyzed by immunofluorescence, western blotting, and quantitative reverse transcription polymerase chain reaction. In vitro, lipopolysaccharide-induced M1 macrophages were incubated separately with Exo-Con and Exo-pre. We performed microRNA (miRNA) sequencing to identify candidate miRNAs and predict their target genes. An miRNA inhibitor was used to confirm the role of miRNAs in macrophage modulation. Exo-pre were more potent than Exo-Con at alleviating DN. Exo-pre administration significantly reduced the number of M1 macrophages and increased the number of M2 macrophages in the kidney compared to Exo-Con administration. Parallel outcomes were observed in the co-culture experiments. Moreover, miR-486-5p was distinctly expressed in Exo-Con and Exo-pre groups, and it played an important role in macrophage polarization by targeting PIK3R1 through the PI3K/Akt pathway. Reducing miR-486-5p levels in Exo-pre abolished macrophage polarization modulation. Exo-pre administration exhibited a superior effect on DN by remodeling the macrophage balance by shuttling miR-486-5p, which targets PIK3R1.
Collapse
Affiliation(s)
- Wanlu Su
- School of Medicine, Nankai University, Tianjin, China
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jian Zhao
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ruofan Hu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haixia Zhang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jia Hu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rui Ren
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yue Zhang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Anning Wang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhaohui Lyu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yiming Mu
- School of Medicine, Nankai University, Tianjin, China
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
31
|
Shalaby AM, Hassan SMA, Abdelnour HM, Alnasser SM, Alorini M, Jaber FA, Alabiad MA, Abdullatif A, Elshaer MMA, Aziz SAMA, Abdelghany EMA. Ameliorative Potential of Bone Marrow-Derived Mesenchymal Stem Cells Versus Prednisolone in a Rat Model of Lung Fibrosis: A Histological, Immunohistochemical, and Biochemical Study. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2024; 30:539-551. [PMID: 38758132 DOI: 10.1093/mam/ozae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/08/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease of unknown origin with limited treatment options and poor prognosis. The encouraging findings from preclinical investigations utilizing mesenchymal stem cells (MSCs) indicated that they could serve as a promising therapeutic alternative for managing chronic lung conditions, such as IPF. The objective of this study was to compare the efficiency of bone marrow-derived MSCs (BM-MSCs) versus prednisolone, the standard anti-inflammatory medication, in rats with bleomycin (BLM)-induced lung fibrosis. Four groups were created: a control group, a BLM group, a prednisolone-treated group, and a BM-MSCs-treated group. To induce lung fibrosis, 5 mg/kg of BLM was administered intratracheally. BLM significantly increased serum levels of pro-inflammatory cytokines and oxidative stress markers. The disturbed lung structure was also revealed by light and transmission electron microscopic studies. Upregulation in the immune expression of alpha-smooth muscle actin, transforming growth factor beta-1, and Bax was demonstrated. Interestingly, all findings significantly regressed on treatment with prednisolone and BM-MSCs. However, treatment with BM-MSCs showed better results than with prednisolone. In conclusion, BM-MSCs could be a promising approach for managing lung fibrosis.
Collapse
Affiliation(s)
- Amany Mohamed Shalaby
- Department of Histology and Cell Biology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Shaimaa Mohamed Abdelfattah Hassan
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shebin El Koum 32511, Egypt
- Department of Anatomy, General Medicine Practice Program, Batterjee Medical College, Aseer 61961, Saudi Arabia
| | - Hanim Magdy Abdelnour
- Department of Medical Biochemistry, Faculty of Human Medicine, Zagazig University, 44519Egypt
| | - Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Mohammed Alorini
- Department of Pathology, College of Medicine, Qassim University, Unaizah 51911, Saudi Arabia
| | - Fatima A Jaber
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Mohamed Ali Alabiad
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Asmaa Abdullatif
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | | | | | - Eman M A Abdelghany
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
32
|
Sun L, Rao S, Kerim K, Lu J, Li H, Zhao S, Shen P, Sun W. A chemically adjustable BMP6-IL6 axis in mesenchymal stem cells drives acute myeloid leukemia cell differentiation. Biochem Pharmacol 2024; 225:116262. [PMID: 38705535 DOI: 10.1016/j.bcp.2024.116262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Chemotherapy alone or in combination with allogeneic stem cell transplantation has been the standard of care for acute myeloid leukemia (AML) for decades. Leukemia relapse with limited treatment options remains the main cause of treatment failure. Therefore, an effective and safe approach to improve treatment outcomes is urgently needed for most AML patients. Mesenchymal stem cells (MSCs) have been reported to efficiently induce apoptosis and shape the fate of acute myeloid leukemia cells. Here, we identified LG190155 as a potent compound that enhances the antileukemia efficiency of MSCs. Pretreatment of MSCs with LG190155 significantly provoked differentiation in both AML patient-derived primary leukemia cells and AML cell lines and reduced the tumor burden in the AML mouse model. Using the quantitative proteomic technique, we discovered a pivotal mechanism that mediates AML cell differentiation, in which autocrine bone morphogenetic protein 6 (BMP6) in MSCs boosted IL-6 secretion and further acted on leukemic cells to trigger differentiation. Furthermore, the activity of the BMP6-IL6 axis was dramatically enhanced by activating vitamin D receptor (VDR) in MSCs. Our data illustrated an effective preactivated approach to reinforcing the antileukemia effect of MSCs, which could serve as an effective therapeutic strategy for AML.
Collapse
Affiliation(s)
- Luchen Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shangrui Rao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kamran Kerim
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jianhua Lu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hongzheng Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shengsheng Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Pingping Shen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Shenzhen Research Institute of NanJing University, Shenzhen 518000, China.
| | - Weijian Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
33
|
Chen H, Wang H, Xu X, Hu Y, Su J, Li D, Li Z, Feng S, Liu J, Zhang H, Wang X. The impact of 5-aminosalicylates on the efficacy of mesenchymal stem cell therapy in a murine model of ulcerative colitis. Int Immunopharmacol 2024; 134:112255. [PMID: 38744176 DOI: 10.1016/j.intimp.2024.112255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Inflammatory bowel disease (IBD) is distinguished by persistent immune-mediated inflammation of the gastrointestinal tract. Previous experimental investigations have shown encouraging outcomes for the use of mesenchymal stem cell (MSC)-based therapy in the treatment of IBD. However, as a primary medication for IBD patients, there is limited information regarding the potential interaction between 5-aminosalicylates (5-ASA) and MSCs. In this present study, we employed the dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mouse model to examine the influence of a combination of MSCs and 5-ASA on the development of UC. The mice were subjected to weight measurement, DAI scoring, assessment of calprotectin expression, and collection of colons for histological examination. The findings revealed that both 5-ASA and MSCs have demonstrated efficacy in the treatment of UC. However, it is noteworthy that 5-ASA exhibits a quicker onset of action, while MSCs demonstrate more advantageous and enduring therapeutic effects. Additionally, the combination of 5-ASA and MSC treatment shows a less favorable efficacy compared to the MSCs alone group. Moreover, our study conducted in vitro revealed that 5-ASA could promote MSC migration, but it could also inhibit MSC proliferation, induce apoptosis, overexpress inflammatory factors (IL-2, IL-12P70, and TNF-α), and reduce the expression of PD-L1 and PD-L2. Furthermore, a significant decrease in the viability of MSCs within the colon was observed as a result of 5-ASA induction. These findings collectively indicate that the use of 5-ASA has the potential to interfere with the therapeutic efficacy of MSC transplantation for the treatment of IBD.
Collapse
MESH Headings
- Animals
- Colitis, Ulcerative/therapy
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/pathology
- Colitis, Ulcerative/chemically induced
- Mesenchymal Stem Cell Transplantation
- Mesalamine/pharmacology
- Mesalamine/therapeutic use
- Disease Models, Animal
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Dextran Sulfate
- Mice
- Humans
- Mice, Inbred C57BL
- Colon/pathology
- Colon/drug effects
- Colon/immunology
- Cells, Cultured
- Male
- Cell Proliferation/drug effects
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
Collapse
Affiliation(s)
- Huanhuan Chen
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - Huimin Wang
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - XiaoJing Xu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, China
| | - Ya'nan Hu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, China
| | - Jing Su
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - Dongdong Li
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - Zimu Li
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - Shixiang Feng
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - Jinming Liu
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China
| | - Huanxiang Zhang
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Xiaoyan Wang
- The Suqian Clinical College of Xuzhou Medical University, 120 Su zhi Road, Sucheng District, Suqian 223800, China.
| |
Collapse
|
34
|
Shimizu Y, Ntege EH, Takahara E, Matsuura N, Matsuura R, Kamizato K, Inoue Y, Sowa Y, Sunami H. Adipose-derived stem cell therapy for spinal cord injuries: Advances, challenges, and future directions. Regen Ther 2024; 26:508-519. [PMID: 39161365 PMCID: PMC11331855 DOI: 10.1016/j.reth.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Spinal cord injury (SCI) has limited treatment options for regaining function. Adipose-derived stem cells (ADSCs) show promise owing to their ability to differentiate into multiple cell types, promote nerve cell survival, and modulate inflammation. This review explores ADSC therapy for SCI, focusing on its potential for improving function, preclinical and early clinical trial progress, challenges, and future directions. Preclinical studies have demonstrated ADSC transplantation's effectiveness in promoting functional recovery, reducing cavity formation, and enhancing nerve regrowth and myelin repair. To improve ADSC efficacy, strategies including genetic modification and combination with rehabilitation are being explored. Early clinical trials have shown safety and feasibility, with some suggesting motor and sensory function improvements. Challenges remain for clinical translation, including optimizing cell survival and delivery, determining dosing, addressing tumor formation risks, and establishing standardized protocols. Future research should focus on overcoming these challenges and exploring the potential for combining ADSC therapy with other treatments, including rehabilitation and medication.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Eisaku Takahara
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Rikako Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Kota Kamizato
- Department of Anesthesiology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| |
Collapse
|
35
|
Joshi R, Suryawanshi T, Mukherjee S, Shukla S, Majumder A. Chromatin Condensation Delays Senescence in Human Mesenchymal Stem Cells by Safeguarding Nuclear Damages during In Vitro Expansion. J Tissue Eng Regen Med 2024; 2024:1543849. [PMID: 40225747 PMCID: PMC11919206 DOI: 10.1155/2024/1543849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/30/2024] [Accepted: 04/05/2024] [Indexed: 04/15/2025]
Abstract
Human mesenchymal stem cells (hMSCs) are multipotent cells that differentiate into adipocytes, chondrocytes, and osteoblasts. Owing to their differentiation potential, hMSCs are among the cells most frequently used for therapeutic applications in tissue engineering and regenerative medicine. However, the number of cells obtained through isolation alone is insufficient for hMSC-based therapies and basic research, which necessitates in vitro expansion. Conventionally, this is often performed on rigid surfaces such as tissue culture plates (TCPs). However, during in vitro expansion, hMSCs lose their proliferative ability and multilineage differentiation potential, rendering them unsuitable for clinical use. Although multiple approaches have been attempted to maintain hMSC stemness during prolonged expansion, finding a suitable culture system remains an unmet need. Recently, a few research groups have shown that hMSCs maintain their stemness over long passages when cultured on soft substrates. In addition, it has been shown that hMSCs cultured on soft substrates have more condensed chromatin and lower levels of histone acetylation compared to those cultured on stiff substrates. Furthermore, it has also been shown that condensing/decondensing chromatin by deacetylation/acetylation can delay replicative senescence in hMSCs during long-term expansion on TCPs. However, the mechanism by which chromatin condensation/decondensation influences nuclear morphology and DNA damage, which are strongly related to the onset of senescence, remains unknown. To answer this question, we cultured hMSCs for long duration in the presence of epigenetic modifiers, histone acetyltransferase inhibitor (HATi), which promotes chromatin condensation by preventing histone acetylation, and histone deacetylase inhibitor (HDACi), which promotes chromatin decondensation, and investigated their effects on various nuclear markers related to senescence. We found that consistent acetylation causes severe nuclear abnormalities, whereas chromatin condensation by deacetylation helps to safeguard the nucleus from damage caused by in vitro expansion.
Collapse
Affiliation(s)
- Rohit Joshi
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Tejas Suryawanshi
- Centre for Research in Nano Technology and Science, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sourav Mukherjee
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shobha Shukla
- Department of Metallurgical Engineering and Materials Science, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
36
|
Zhang Y, Zhang Y, Hu A, Meng F, Cui P, Li T, Cui G. Mesenchymal stem cells derived from CHIR99021 and TGF‑β induction remained on the colicomentum and improved cardiac function of a rat model of acute myocardium infarction. Exp Ther Med 2024; 27:182. [PMID: 38515646 PMCID: PMC10952379 DOI: 10.3892/etm.2024.12470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/17/2024] [Indexed: 03/23/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have been regarded as a potential stem cell source for cell therapy. However, the production of cells with mesenchymal potential from hiPSCs through spontaneous differentiation is time consuming and laborious. In the present study, the combined use of the GSK-3 inhibitor CHIR99021 and TGF-β was used to obtain mesenchymal stem cell (MSC)-like cells from hiPSCs. During the induction process, the transcription of epithelial-mesenchymal transition (EMT)-related genes N-cadherin and Vimentin in the transformed cells was upregulated, whereas the transcription of E-cadherin and pluripotency-related transcription factors SOX2, OCT4 and NANOG did not change significantly. This indicated that whilst cells were pluripotent, EMT was initiated by the upregulation of transcription of EMT promoting genes. Both SMAD-dependent and independent signalling pathways were significantly activated by the combined induction treatment compared with the single factor induction. The hiPSC-derived MSC-like cells (hiPSC-MSCs) expressed MSC-related markers and acquired osteogenic, chondrogenic and adipogenic differentiation potentials. After being injected into the peritoneal cavity of rats, the hiPSC-MSCs secreted angiogenic and immune-regulatory factors and remained on the colicomentum for 3 weeks. Within an 11-week period, four intraperitoneal hiPSC-MSC injections (1x107 cells/injection) into acute myocardial infarction (AMI) model rats significantly increased the left ventricular ejection fraction, left ventricular fractional shortening and angiogenesis and significantly reduced scar size and the extent of apoptosis in the infarcted area compared with that of the control PBS injection. Symptoms of hiPSC-MSC-induced immune reaction or tumour formation were not observed over the course of the experiment in the hiSPC-MSC treated rats. In conclusion, the CHIR99021 and TGF-β combined induction was a rapid and effective method to obtain MSC-like cells from hiPSCs and multiple high dose intraperitoneal injections of hiPSC-derived MSCs were safe and effective at restoring cardiac function in an AMI rat model.
Collapse
Affiliation(s)
- Yusen Zhang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yanmin Zhang
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Azhen Hu
- Shenzhen Key Laboratory of Drug Addiction and Safe Medication, Shenzhen PKU-HKUST Medical Centre, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Fanhua Meng
- Reproductive Medical Centre, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Peng Cui
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Tianshi Li
- Department of Plastic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Guanghui Cui
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
37
|
Ozkan S, Isildar B, Sahin H, Saygi HI, Konukoglu D, Koyuturk M. Comparative analysis of effects of conditioned mediums obtained from 2D or 3D cultured mesenchymal stem cells on kidney functions of diabetic rats: Early intervention could potentiate transdifferentiation of parietal epithelial cell into podocyte precursors. Life Sci 2024; 343:122543. [PMID: 38460812 DOI: 10.1016/j.lfs.2024.122543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
AIM The secretome of mesenchymal stem cells (MSCs) could be a potential therapeutic intervention for diabetes and associated complications like nephropathy. This study aims to evaluate the effects of conditioned mediums (CMs) collected from umbilical cord-derived MSCs incubated under 2-dimensional (2D) or 3D culture conditions on kidney functions of rats with type-I diabetes (T1D). MAIN METHODS Sprague-Dawley rats were treated with 20 mg/kg streptozocin for 5 consecutive days to induce T1D, and 12 doses of CMs were applied intraperitoneally for 4 weeks. The therapeutic effects of CMs were comparatively investigated by biochemical, physical, histopathological, and immunohistochemical analysis. KEY FINDINGS 3D-CM had significantly higher total protein concentration than the 2D-CM Albumin/creatinine ratios of both treatment groups were significantly improved in comparison to diabetes. Light microscopic evaluations showed that glomerular and cortical tubular damages were significantly ameliorated in only the 3D-CM applied group compared to the diabetes group, which were correlated with transmission electron microscopic observations. The nephrin and synaptopodin expressions increased in both treatment groups compared to diabetes. The WT1, Ki-67, and active caspase-3 expressions in glomeruli and parietal layers of the treatment groups suggest that both types of CMs suppress apoptosis and promote possible parietal epithelial cells' (PECs') transdifferentiation towards podocyte precursor cells by switching on WT1 expression in parietal layer rather than inducing new cell proliferation. SIGNIFICANCE 3D-CM was found to be more effective in improving kidney functions than 2D-CM by ameliorating glomerular damage through the possible mechanism of transdifferentiation of PECs into podocyte precursors and suppressing glomerular apoptosis.
Collapse
Affiliation(s)
- Serbay Ozkan
- Izmir Katip Çelebi University, Faculty of Medicine, Histology and Embryology Department, Turkey; Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Turkey
| | - Basak Isildar
- Balıkesir University, Faculty of Medicine, Histology and Embryology Department, Turkey; Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Turkey
| | - Hakan Sahin
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Turkey
| | - Halil Ibrahim Saygi
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Turkey; Istanbul Medeniyet University, Faculty of Medicine, Histology and Embryology Department, Turkey
| | - Dildar Konukoglu
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Medical Biochemistry Department, Turkey
| | - Meral Koyuturk
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Histology and Embryology Department, Turkey.
| |
Collapse
|
38
|
Xu Q, Xiao Z, Yang Q, Yu T, Deng X, Chen N, Huang Y, Wang L, Guo J, Wang J. Hydrogel-based cardiac repair and regeneration function in the treatment of myocardial infarction. Mater Today Bio 2024; 25:100978. [PMID: 38434571 PMCID: PMC10907859 DOI: 10.1016/j.mtbio.2024.100978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
A life-threatening illness that poses a serious threat to human health is myocardial infarction. It may result in a significant number of myocardial cells dying, dilated left ventricles, dysfunctional heart function, and ultimately cardiac failure. Based on the development of emerging biomaterials and the lack of clinical treatment methods and cardiac donors for myocardial infarction, hydrogels with good compatibility have been gradually applied to the treatment of myocardial infarction. Specifically, based on the three processes of pathophysiology of myocardial infarction, we summarized various types of hydrogels designed for myocardial tissue engineering in recent years, including natural hydrogels, intelligent hydrogels, growth factors, stem cells, and microRNA-loaded hydrogels. In addition, we also describe the heart patch and preparation techniques that promote the repair of MI heart function. Although most of these hydrogels are still in the preclinical research stage and lack of clinical trials, they have great potential for further application in the future. It is expected that this review will improve our knowledge of and offer fresh approaches to treating myocardial infarction.
Collapse
Affiliation(s)
- Qiaxin Xu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, 510630, China
| | - Qianzhi Yang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Tingting Yu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Xiujiao Deng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Nenghua Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Lihong Wang
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Endocrinology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jun Guo
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
39
|
Sirpilla O, Sakemura RL, Hefazi M, Huynh TN, Can I, Girsch JH, Tapper EE, Cox MJ, Schick KJ, Manriquez-Roman C, Yun K, Stewart CM, Ogbodo EJ, Kimball BL, Mai LK, Gutierrez-Ruiz OL, Rodriguez ML, Gluscevic M, Larson DP, Abel AM, Wierson WA, Olivier G, Siegler EL, Kenderian SS. Mesenchymal stromal cells with chimaeric antigen receptors for enhanced immunosuppression. Nat Biomed Eng 2024; 8:443-460. [PMID: 38561490 PMCID: PMC12080371 DOI: 10.1038/s41551-024-01195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Allogeneic mesenchymal stromal cells (MSCs) are a safe treatment option for many disorders of the immune system. However, clinical trials using MSCs have shown inconsistent therapeutic efficacy, mostly owing to MSCs providing insufficient immunosuppression in target tissues. Here we show that antigen-specific immunosuppression can be enhanced by genetically modifying MSCs with chimaeric antigen receptors (CARs), as we show for E-cadherin-targeted CAR-MSCs for the treatment of graft-versus-host disease in mice. CAR-MSCs led to superior T-cell suppression and localization to E-cadherin+ colonic cells, ameliorating the animals' symptoms and survival rates. On antigen-specific stimulation, CAR-MSCs upregulated the expression of immunosuppressive genes and receptors for T-cell inhibition as well as the production of immunosuppressive cytokines while maintaining their stem cell phenotype and safety profile in the animal models. CAR-MSCs may represent a widely applicable therapeutic technology for enhancing immunosuppression.
Collapse
Affiliation(s)
- Olivia Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - R Leo Sakemura
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Mehrdad Hefazi
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Truc N Huynh
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - James H Girsch
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Erin E Tapper
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Michelle J Cox
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Kendall J Schick
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Claudia Manriquez-Roman
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Carli M Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Ekene J Ogbodo
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Brooke L Kimball
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Long K Mai
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Omar L Gutierrez-Ruiz
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Makena L Rodriguez
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Martina Gluscevic
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Daniel P Larson
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | - Alex M Abel
- LifEngine Animal Health Laboratories Incorporated, Rochester, MN, USA
| | - Wesley A Wierson
- LifEngine Animal Health Laboratories Incorporated, Rochester, MN, USA
| | - Gloria Olivier
- Department of Business Development, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
40
|
Dias IE, Dias IR, Franchi-Mendes T, Viegas CA, Carvalho PP. A Comprehensive Exploration of Therapeutic Strategies in Inflammatory Bowel Diseases: Insights from Human and Animal Studies. Biomedicines 2024; 12:735. [PMID: 38672091 PMCID: PMC11048724 DOI: 10.3390/biomedicines12040735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a collective term for a group of chronic inflammatory enteropathies which are characterized by intestinal inflammation and persistent or frequent gastrointestinal signs. This disease affects more than 3.5 million humans worldwide and presents some similarities between animal species, in particular, dogs and cats. Although the underlying mechanism that triggers the disease is not yet well understood, the evidence suggests a multifactorial etiology implicating genetic causes, environmental factors, microbiota imbalance, and mucosa immune defects, both in humans and in dogs and cats. Conventional immunomodulatory drug therapies, such as glucocorticoids or immunosuppressants, are related with numerous adverse effects that limit its long-term use, creating the need to develop new therapeutic strategies. Mesenchymal stromal cells (MSCs) emerge as a promising alternative that attenuates intestinal inflammation by modulating inflammatory cytokines in inflamed tissues, and also due to their pro-angiogenic, anti-apoptotic, anti-fibrotic, regenerative, anti-tumor, and anti-microbial potential. However, this therapeutic approach may have important limitations regarding the lack of studies, namely in veterinary medicine, lack of standardized protocols, and high economic cost. This review summarizes the main differences and similarities between human, canine, and feline IBD, as well as the potential treatment and future prospects of MSCs.
Collapse
Affiliation(s)
- Inês Esteves Dias
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; (I.E.D.); (I.R.D.)
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Isabel Ribeiro Dias
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; (I.E.D.); (I.R.D.)
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, Quinta de Prados, 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- CECAV—Centre for Animal Sciences and Veterinary Studies, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- AL4AnimalS—Associate Laboratory for Animal and Veterinary Sciences, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Teresa Franchi-Mendes
- Department of Bioengineering and IBB—Institute for Bioengineering and Biosciences at Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carlos Antunes Viegas
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- CECAV—Centre for Animal Sciences and Veterinary Studies, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- AL4AnimalS—Associate Laboratory for Animal and Veterinary Sciences, Quinta de Prados, 5000-801 Vila Real, Portugal
- CIVG—Vasco da Gama Research Center, University School Vasco da Gama (EUVG), Campus Universitário, Av. José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal;
| | - Pedro Pires Carvalho
- CIVG—Vasco da Gama Research Center, University School Vasco da Gama (EUVG), Campus Universitário, Av. José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal;
- Vetherapy—Research and Development in Biotechnology, 3020-210 Coimbra, Portugal
| |
Collapse
|
41
|
Ballikaya E, Babadag S, San Keskin NO, Çelebi-Saltik B. The Effects of Grape Seed Oligomeric Proanthocyanidin and Nisin on Dental Pulp Stem Cells. Acta Stomatol Croat 2024; 58:2-17. [PMID: 38562220 PMCID: PMC10981911 DOI: 10.15644/asc58/1/1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/09/2024] [Indexed: 04/04/2024] Open
Abstract
Objective This study aimed to evaluate the biological effects of "proanthocyanidin" (PA), and "nisin" (Ni), on dental pulp stem cells (DPSCs) and LPS-induced DPSCs as well as their antimicrobial effects against S. aureus and E. coli. Materials and methods After characterization of DPSCs, cytotoxicity of PA and Ni on DPSCs were evaluated using a water-soluble tetrazolium salt (WST-1). The cytokines and chemokines released by DPSCs and the expression levels of IL-6, IL-8, and TNF alpha were detected with human Cytokine Array C5 and enzyme-linked immunosorbent assay (ELİSA), respectively. The antibacterial activities of PA and Ni were tested using the drop plate method. Results PA at 75 μg/ml increased cell viability, decreased TNF-α expression of DPSCs, did not show any cytotoxic effects on LPS-induced DPSCs, and also showed a tendency to decrease TNF-α expression. PA at 75 μg/ml exhibited higher expressions of TIMP-2, OPG, IL-7, and IL-8 in LPS-induced DPSCs compared to DPSCs. Ni at 100 μg/ml decreased TNF-α expression in DPSCs with no cytotoxic effects. It provided increased cell viability and a downregulation trend of TNF-α expression in LPS-induced DPSCs. Both Ni and PA provided strong antibacterial effects against S. aureus. Ni at 200μg/ml had strong antibacterial effects against E. coli without affecting negatively the viability of both DPSCs and LPS-induced DPSCs and showed anti-inflammatory activity by decreasing TNF-α expression. PA provided strong antibacterial effects against E. coli at 200 μg/ml but affected DPSCs viability negatively. Conclusion PA and Ni at specific concentrations exhibited immunomodulatory activity on DPSCs and LPS-induced DPSCs without any cytotoxic effects and strong antibacterial effects on S. aureus.
Collapse
Affiliation(s)
- Elif Ballikaya
- Department of Oral and Dental Health Research, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Department of Pediatric Dentistry, Hacettepe University Faculty of Dentistry, Ankara, Turkey
| | - Sena Babadag
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Nalan Oya San Keskin
- Polatlı Science and Literature Faculty, Nanosan Laboratory, Ankara Hacı Bayram Veli University, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Oral and Dental Health Research, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
42
|
Cabello-Arista B, Melgarejo-Ramírez Y, Retana-Flores A, Martínez-López V, Márquez-Gutiérrez E, Almanza-Pérez J, Lecona H, Reyes-Frías ML, Ibarra C, Martínez-Pardo ME, Velasquillo C, Sánchez-Sánchez R. Effects of mesenchymal stem cell culture on radio sterilized human amnion or radio sterilized pig skin in burn wound healing. Cell Tissue Bank 2024; 25:255-267. [PMID: 35059955 DOI: 10.1007/s10561-021-09976-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 11/02/2021] [Indexed: 11/29/2022]
Abstract
Deep second and third degree burns treatment requires fibroblasts, keratinocytes and other skin cells in order to grow new dermis and epidermis. Cells can proliferate, secrete growth factors and extracellular matrix required to repair the damaged tissue. Radiosterilized human amnion and radiosterilized pig skin have been used as natural origin skin dressings for burned patients. Adipose-derived mesenchymal stem cells can differentiate into fibroblasts and keratinocytes and improve wound-healing progress. These cells can stimulate vascular tissue formation, release growth factors, synthetize new extracellular matrix and immunoregulate other cells. In this study, we developed mesenchymal stem cells-cellularized skin substitutes based from radiosterilized human amnion or pig skin. Third-degree burns were induced in mice animal models to evaluate the effect of cellularized skin substitutes on burn wound healing. Mesenchymal phenotype was immunophenotypically confirmed by flow cytometry and cell viability was close to 100%. Skin recovery was evaluated in burned mice after seven and fourteen days post-coverage with cellularized and non-cellularized sustitutes. Histological techniques and immunofluorescence were used to evaluate re-epithelization and type I collagen deposition. We determined that cellularized-human amnion or cellularized-pig skin in combination with mesenchymal stem cells improve extracellular matrix deposition. Both cellularized constructs increase detection of type I collagen in newly formed mouse skin and can be potentially used as skin coverage for further clinical treatment of burned patients.
Collapse
Affiliation(s)
- B Cabello-Arista
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - Y Melgarejo-Ramírez
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - A Retana-Flores
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - V Martínez-López
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - E Márquez-Gutiérrez
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - J Almanza-Pérez
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S.,, Universidad Autónoma Metropolitana-Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - H Lecona
- Bioterio y Cirugía Experimental, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - M L Reyes-Frías
- Banco de Tejidos Radioesterilizados, Instituto Nacional de Investigaciones Nucleares (BTR-ININ), Carretera México-Toluca S/N La Marquesa, C.P. 52750, Ocoyoacac, Edo. Mex, Mexico
| | - C Ibarra
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - M E Martínez-Pardo
- Banco de Tejidos Radioesterilizados, Instituto Nacional de Investigaciones Nucleares (BTR-ININ), Carretera México-Toluca S/N La Marquesa, C.P. 52750, Ocoyoacac, Edo. Mex, Mexico
| | - C Velasquillo
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico
| | - R Sánchez-Sánchez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, C.P. 14389, Mexico City, Mexico.
- Escuela de Ingeniería y Ciencias, Departamento de Bioingeniería, Instituto Tecnológico de Monterrey, Puente 222, Col. Arboledas del Sur, C.P. 14380, Mexico City, Mexico.
| |
Collapse
|
43
|
Pires F, Silva JC, Ferreira FC, Portugal CAM. Heparinized Acellular Hydrogels for Magnetically Induced Wound Healing Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9908-9924. [PMID: 38381140 DOI: 10.1021/acsami.3c18877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The control of angiogenesis has the potential to be used for regulation of several pathological and physiological processes, which can be instrumental on the development of anticancer and wound healing therapeutical approaches. In this study, mesenchymal stem/stromal cells (MSCs) were seeded on magnetic-responsive gelatin, with or without heparin functionalization, and exposed to a static 0.08 T magnetic field (MF), for controlling their anti-inflammatory and angiogenic activity, with the aim of accelerating tissue healing. For the first time, it was examined how the amount of heparin and magnetic nanoparticles (MNPs) distributed on gelatin scaffolds affected the mechanical properties of the hydrogels and the morphology, proliferation, and secretome profiling of MSCs. The findings demonstrated that the addition of MNPs and heparin affects the hydrogel swelling capacity and renders distinct MSC proliferation rates. Additionally, MF acts as a topographical cue to guide MSCs alignment and increases the level of expression of specific genes and proteins that promote angiogenesis. The results also suggested that the presence of higher amounts of heparin (10 μg/cm3) interferes with the secretion and limits the capacity of angiogenic factors to diffuse through the hydrogel and into the culture medium. Ultimately, this study shows that acellular heparinized hydrogels efficiently retain the angiogenic growth factors released by magnetically stimulated MSCs thus rendering superior wound contraction (55.8% ± 0.4%) and cell migration rate (49.4% ± 0.4%), in comparison to nonheparinized hydrogels (35.2% ± 0.7% and 37.8% ± 0.7%, respectively). Therefore, these heparinized magnetic hydrogels can be used to facilitate angiogenesis in various forms of tissue damage including bone defects, skin wounds, and cardiovascular diseases, leading to enhanced tissue regeneration.
Collapse
Affiliation(s)
- Filipa Pires
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - João Carlos Silva
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carla A M Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
44
|
Kheder RK, Darweesh O, Hussen BM, Abdullah SR, Basiri A, Taheri M. Mesenchymal stromal cells (MSCs) as a therapeutic agent of inflammatory disease and infectious COVID-19 virus: live or dead mesenchymal? Mol Biol Rep 2024; 51:295. [PMID: 38340168 DOI: 10.1007/s11033-023-09174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/12/2024]
Abstract
The COVID-19 infection is a worldwide disease that causes numerous immune-inflammatory disorders, tissue damage, and lung dysfunction. COVID-19 vaccines, including those from Pfizer, AstraZeneca, and Sinopharm, are available globally as effective interventions for combating the disease. The severity of COVID-19 can be most effectively reduced by mesenchymal stromal cells (MSCs) because they possess anti-inflammatory activity and can reverse lung dysfunction. MSCs can be harvested from various sources, such as adipose tissue, bone marrow, peripheral blood, inner organs, and neonatal tissues. The regulation of inflammatory cytokines is crucial in inhibiting inflammatory diseases and promoting the presence of anti-inflammatory cytokines for infectious diseases. MSCs have been employed as therapeutic agents for tissue damage, diabetes, autoimmune diseases, and COVID-19 patients. Our research aimed to determine whether live or dead MSCs are more suitable for the treatment of COVID-19 patients. Our findings concluded that dead MSCs, when directly administered to the patient, offer advantages over viable MSCs due to their extended presence and higher levels of immune regulation, such as T-reg, B-reg, and IL-10, compared to live MSCs. Additionally, dead and apoptotic MSCs are likely to be more readily captured by monocytes and macrophages, prolonging their presence compared to live MSCs.
Collapse
Affiliation(s)
- Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Sulaymaniyah, Iraq
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Omeed Darweesh
- College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, Erbil, 44001, Iraq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Scineces, Tehran, Iran
| | - Mohammad Taheri
- Institue of Human Genetics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
45
|
Alavi-Dana SMM, Gholami Y, Meghdadi M, Fadaei MS, Askari VR. Mesenchymal stem cell therapy for COVID-19 infection. Inflammopharmacology 2024; 32:319-334. [PMID: 38117433 DOI: 10.1007/s10787-023-01394-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023]
Abstract
COVID-19 emerged in December 2019 in Wuhan, China, spread worldwide rapidly, and caused millions of deaths in a short time. Many preclinical and clinical studies were performed to discover the most efficient therapy to reduce the mortality of COVID-19 patients. Among various approaches for preventing and treating COVID-19, mesenchymal stem cell (MSC) therapy can be regarded as a novel and efficient treatment for managing COVID-19 patients. In this review, we explain the pathogenesis of COVID-19 infection in humans and discuss the role of MSCs in suppressing the inflammation and cytokine storm produced by COVID-19. Then, we reviewed the clinical trial and systematic review studies that investigated the safety and efficacy of MSC therapy in the treatment of COVID-19 infection.
Collapse
Affiliation(s)
| | - Yazdan Gholami
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Meghdadi
- Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Saleh Fadaei
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Vahid Reza Askari
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran.
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
46
|
Ha GH, Yeon JY, Kim KH, Lee DM, Chae HY, Nam H, Lee K, Kim DO, Kim CK, Joo KM. Thrombin Priming Promotes the Neuroprotective Effects of Human Wharton's Jelly-Derived Mesenchymal Stem Cells Via the HGF/AKT/STAT3 Signaling Pathway. Stem Cells Dev 2024; 33:89-103. [PMID: 38164089 DOI: 10.1089/scd.2023.0191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Mesenchymal stem cells (MSCs) directly differentiate into neurons and endothelial cells after transplantation, and their secretome has considerable potential for treating brain injuries. Previous studies have suggested that the effects of MSCs priming with exposure to hypoxia, cytokines, growth factors, or chemical agents could optimize the paracrine potency and therapeutic potential of MSCs. Studies have suggested that thrombin-primed Wharton's Jelly-derived mesenchymal stem cells (Th.WJ-MSCs) significantly enhance the neuroprotective beneficial effects of naive MSCs in brain injury such as hypoxic-ischemic brain injury (HIE) and intraventricular hemorrhage (IVH). This study aimed to characterize WJ-MSCs in terms of stem cell markers, differentiation, cell proliferation, and paracrine factors by comparing naive and Th.WJ-MSCs. We demonstrated that compared with naive MSCs, Th.MSCs significantly enhanced the neuroprotective effects in vitro. Moreover, we identified differentially expressed proteins in the conditioned media of naive and Th.WJ-MSCs by liquid chromatography-tandem mass spectrometry analysis. Secretome analysis of the conditioned medium of WJ-MSCs revealed that such neuroprotective effects were mediated by paracrine effects with secretomes of Th.WJ-MSCs, and hepatocyte growth factor was identified as a key paracrine mediator. These results can be applied further in the preclinical and clinical development of effective and safe cell therapeutics for brain injuries such as HIE and IVH.
Collapse
Affiliation(s)
- Geun-Hyoung Ha
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Je Young Yeon
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ki Hoon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Du Man Lee
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Hye Yun Chae
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Hyun Nam
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kyunghoon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Dong Oh Kim
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Medical Innovation Technology, Inc. (MEDINNO, Inc.), Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
47
|
Lee J, Nguyen S, Bhattacharya S. Optic nerve regeneration: Potential treatment approaches. Curr Opin Pharmacol 2024; 74:102428. [PMID: 38171063 PMCID: PMC10922496 DOI: 10.1016/j.coph.2023.102428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024]
Abstract
The optic nerve, predominantly constituted by the axons of retinal ganglion cells (RGCs), lacks the ability to regenerate and re-establish function after injury. RGCs are crucial for visual function, and thus, RGC death contributes to the development of numerous progressive neurodegenerative optic neuropathies including glaucoma, ischemic optic neuropathy, and optic neuritis. Regenerating optic nerve axons poses numerous challenges due to factors such as the intricate and inhibitory conditions that exist within their environment, intrinsic breaks to regeneration, and the geometric tortuosity that offers physical hindrance to axon growth. However, recent research advancements offer hope for clinically meaningful regeneration for those who suffer from optic nerve damage. In this review, we highlight the current treatment approaches for optic nerve axon regeneration.
Collapse
Affiliation(s)
- Jessica Lee
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, USA; College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
| | - Sherilyn Nguyen
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, USA; College of Osteopathic Medicine, Nova Southeastern University, Tampa, FL, USA
| | - Sanjoy Bhattacharya
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, USA.
| |
Collapse
|
48
|
Nejabati HR, Nikzad S, Roshangar L. Therapeutic Potential of Mesenchymal Stem Cells in PCOS. Curr Stem Cell Res Ther 2024; 19:134-144. [PMID: 37198984 DOI: 10.2174/1574888x18666230517123256] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a major reproductive endocrine disorder affecting different facets of a woman's life, comprising reproduction, metabolism, and mental health. Recently, several research groups have brought attention to the therapeutic capacity of mesenchymal stem cells (MSCs) for the treatment of female reproductive disorders. It is highlighted that the treatment with bone marrow mesenchymal stem cells (BMMSCs) considerably diminishes the levels of some inflammatory markers as well as essential genes for ovarian production of androgens, which are considerably higher in theca cells of PCOS women than in those of healthy cases. In addition, studies show that BMMSCs improve in vitro maturation (IVM) of germinal vesicles (GVs) and the number of antral follicles while lessening the number of primary and preantral follicles in mice with PCOS compared to healthy controls. Regarding adipose- derived mesenchymal stem cells (AdMSCs), these cells restore the ovarian structure, enhance the number of oocytes and corpora luteum, and diminish the number of aberrant cystic follicles in PCOS rats. Some research also indicates that umbilical cord mesenchymal stem cells (UC-MSCs) alleviate the inflammation of granulosa cells in women with PCOS. Therefore, due to the limited research on MSC therapy in PCOS, in this review, we summarize the current knowledge on the therapeutic potential of three types of MSCs: BMMSCs, AdMSCs, UC-MSCs and their secretome in the treatment of PCOS.
Collapse
Affiliation(s)
- Hamid Reza Nejabati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadeneh Nikzad
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Mani A, Hotra J, Blackwell SC, Goetzl L, Refuerzo JS. Mesenchymal Stem Cells Suppress Inflammatory Cytokines in Lipopolysaccharide Exposed Preterm and Term Human Pregnant Myometrial Cells. AJP Rep 2024; 14:e69-e73. [PMID: 38370325 PMCID: PMC10874693 DOI: 10.1055/a-2216-9194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/19/2023] [Indexed: 02/20/2024] Open
Abstract
Objective The objective of this study was to determine the cytokine response in human pregnant preterm and term myometrial cells exposed to lipopolysaccharide (LPS) and cocultured with mesenchymal stem cells (MSCs). Study Design Myometrium was obtained at cesarean delivery in term and preterm patients. Human myometrial cells were exposed to 5 μg/mL LPS for 4 hours followed by 1 μg/mL LPS for 24 hours and were cocultured with MSCs for 24 hours. Culture supernatants were collected at 24 hours and expression of cytokines, including interleukin-1β (IL-1β), IL-6, IL-8, tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), and IL-10, was quantified by enzyme-linked immunosorbent assay. Results There was significantly increased expression of the proinflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α in preterm myometrial cells treated with LPS compared with untreated preterm myometrial cells. Coculture with MSCs significantly suppressed the proinflammatory cytokine levels in LPS-treated preterm versus treated term myometrial cells. Moreover, MSC cocultured preterm myometrial cells expressed increased levels of the anti-inflammatory cytokines TGF-β and IL-10 compared with treated term myometrial cells. Conclusion MSCs ameliorate LPS-mediated inflammation in preterm human myometrial cells compared with term myometrial cells. Immunomodulatory effects of MSCs mediated through anti-inflammatory cytokine regulation suggest a potential cell-based therapy for preterm birth.
Collapse
Affiliation(s)
- Arunmani Mani
- Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| | - John Hotra
- Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| | - Sean C. Blackwell
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Laura Goetzl
- Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| | - Jerrie S. Refuerzo
- Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
50
|
Cates WT, Denbeigh JM, Salvagno RT, Kakar S, van Wijnen AJ, Eaton C. Inflammatory Markers Involved in the Pathogenesis of Dupuytren's Contracture. Crit Rev Eukaryot Gene Expr 2024; 34:1-35. [PMID: 38912961 DOI: 10.1615/critreveukaryotgeneexpr.2024052889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Dupuytren's disease is a common fibroproliferative disease that can result in debilitating hand deformities. Partial correction and return of deformity are common with surgical or clinical treatments at present. While current treatments are limited to local procedures for relatively late effects of the disease, the pathophysiology of this connective tissue disorder is associated with both local and systemic processes (e.g., fibrosis, inflammation). Hence, a better understanding of the systemic circulation of Dupuytren related cytokines and growth factors may provide important insights into disease progression. In addition, systemic biomarker analysis could yield new concepts for treatments of Dupuytren that attenuate circulatory factors (e.g., anti-inflammatory agents, neutralizing antibodies). Progress in the development of any disease modifying biologic treatment for Dupuytren has been hampered by the lack of clinically useful biomarkers. The characterization of nonsurgical Dupuytren biomarkers will permit disease staging from diagnostic and prognostic perspectives, as well as allows evaluation of biologic responses to treatment. Identification of such markers may transcend their use in Dupuytren treatment, because fibrotic biological processes fundamental to Dupuytren are relevant to fibrosis in many other connective tissues and organs with collagen-based tissue compartments. There is a wide range of potential Dupuytren biomarker categories that could be informative, including disease determinants linked to genetics, collagen metabolism, as well as immunity and inflammation (e.g., cytokines, chemokines). This narrative review provides a broad overview of previous studies and emphasizes the importance of inflammatory mediators as candidate circulating biomarkers for monitoring Dupuytren's disease.
Collapse
Affiliation(s)
- William T Cates
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Janet M Denbeigh
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | | |
Collapse
|